Abstract

Traditionally, many invasive fungal infections were associated with a poor prognosis, because effective therapeutic options were limited. The recent development of new antifungal agents has significantly contributed to the successful treatment of fungal diseases. These drugs offer novel mechanisms of action and expanded spectrums of activity over traditional treatment options. However, with these new agents comes the need for increased awareness of the potential interactions and toxicities associated with these drugs. Therefore, an understanding of the pharmacokinetic and pharmacodynamic properties of the classes of antifungal compounds is vital for the effective management of invasive fungal infections. This review provides a summary of the pharmacologic principles involved in treatment of fungal diseases.

The number of agents available to treat invasive fungal infections has increased by 30% since the turn of the millennium. Although that statistic is impressive, it brings the total number of approved systemic antifungal drugs to only 14 [1], with the potential for 1 more product to possibly emerge this year. These recent additions have provided clinicians with a tool previously lacking in the management of these life-threatening infections: therapeutic alternatives.

Along with new options, however, comes the need to understand the uniqueness of each agent, including its role in therapy, toxicity profile, and interactions with concomitant medications. To attain the maximum effect from these agents, clinicians should also become familiar with strategies to optimize efficacy through an understanding of pharmacokinetic and pharmacodynamic properties. These characteristics are unique for each class of antifungal drug and even for each member within a class. In many cases, this variability is not subtle and merits careful attention.

An additional concern related to the increasing number of antifungal drugs is the rapid increase in expenditures associated with their use. Many institutions throughout the United States are struggling with the increased financial burden related to the prescribing of these antifungal drugs [2]. Optimization of therapies through targeted application of various kinetic and dynamic principles may be one strategy to maximize the cost-effectiveness of treatment of invasive fungal infections. This review focuses on the pharmacologic principles involved in treatment of fungal disease and compares and contrasts the differences among the available agents. Given its role as an agent used primarily to treat superficial infections, terbinafine will not be included in this discussion.

History And Mechanisms of Action

Amphotericin B has been the mainstay of antifungal therapy since its release in the 1950s [3]. This agent emerged as the preferred polyene over the more toxic agent in this class, nystatin. Nystatin has since been relegated to topical and localized therapy because of its unfavorable adverse effect profile. The polyene agents exert their antifungal activity via binding to ergosterol in the fungal cell membrane (figure 1). This disrupts cell permeability and results in rapid cell death [4]. To date, amphotericin B remains the broadest-spectrum antifungal agent available, with activity against many clinically relevant yeasts and moulds (table 1). During the 1990s, newer lipid preparations of amphotericin B, including amphotericin B lipid complex (Abelcet; Enzon), liposomal amphotericin B (AmBisome; Astellas Pharma US), and amphotericin B colloidal dispersion (Amphotec; Three Rivers Pharmaceuticals) were developed to alleviate drug toxicity [27]. These agents possess the same spectrum of activity as does amphotericin B deoxycholate. Each agent has been shown to decrease nephrotoxicity in comparison with the conventional preparation of amphotericin B [28]. However, with the exception of findings for histoplasmosis, data supporting increased efficacy of the lipid products against common opportunistic fungal pathogens are lacking [29].

Figure 1

Targets of systemic antifungal agents

Amphotericin B and griseofulvin remained the only systemic therapeutic options for invasive fungal disease until the early 1970s, when flucytosine (Ancobon; Valeant Pharmaceuticals) was released. Flucytosine is a pyrimidine analog that exerts antifungal activity via inhibition of both DNA synthesis and protein synthesis in the fungal cell [30]. It also holds the distinction of being part of the only routinely recommended combination antifungal regimen for treatment of cryptococcal meningitis [31]. Unfortunately, the toxicity of this agent and the rapid development of resistance when used as monotherapy have precluded its routine use for the treatment of other invasive infections [5, 32, 33].

In 1979, the first systemic azole antifungal agent, ketoconazole, was introduced [4]. Azole agents exert their antifungal activity by blocking the demethylation of lanosterol, thereby inhibiting ergosterol synthesis [4]. Ketoconazole was followed chronologically by fluconazole (Diflucan; Pfizer), itraconazole (Sporanox; Janssen Pharmaceuticals), and voriconazole (Vfend; Pfizer) [34]. Four investigational agents remain in development: posaconazole, which has been submitted for US Food and Drug Administration approval, and ravuconazole, BAL8557, and albaconazole, which remain under study. Each agent offers a specific antifungal spectrum. Earlier agents in the class demonstrated potent activity against some, but not all, yeasts, and itraconazole had some activity against moulds, including Aspergillus species (table 1). The newer, expanded-spectrum triazoles have been shown to have cidal activity against a wide spectrum of moulds, as well as enhanced activity against Candida species and other yeasts [6, 7, 35].

The echinocandins represent the newest class of antifungals. Caspofungin (Cancidas; Merck) was released in 2001. This was followed by micafungin (Mycamine; Astellas Pharma US) in 2005 and anidulafungin (Eraxis; Pfizer). The mechanism of activity of the echinocandins is inhibition of the production of (1→3)-β-D-glucan, an essential component in the fungal cell wall [4]. The spectrum of activity is therefore limited to pathogens that rely on these glucan polymers and is less broad than the spectrums of the polyene or azole agents. The echinocandins exhibit fungicidal activity against many Candida species, making this drug class a desirable alternative to the azole agents, which exhibit only static activity against yeasts [35, 36]. Because mammalian cells have no cell wall, the echinocandins have very few toxic adverse effects in humans.

Pharmacokinetics and Pharmacodynamics

The selection of an appropriate antifungal agent depends on multiple factors in addition to the spectrum of activity. As with antibacterial therapy, the routes of administration and elimination are often important considerations in selecting a drug. This is particularly true when the optimal therapy for a patient with a fungal infection is being determined. Alterations in gastrointestinal tract integrity, impaired renal or hepatic function, and limited intravenous access are frequent issues for patients who are at high risk of acquiring fungal disease.

Further complicating the clinical picture is the variability in available formulations among different antifungal agents. Many drugs are available only as intravenous preparations (e.g., amphotericin B preparations and echinocandin agents) or only as oral preparations (e.g., posaconazole and flucytosine) because of differences in solubility and oral bioavailability. For the agents that can be administered by multiple routes (e.g., fluconazole, itraconazole, and voriconazole), there are often difficulties in administration of these preparations because of toxicities, drug interactions, and variability with different product formulations. Therefore, it is important to have an appreciation of the differences among these drugs with regard to their pharmacokinetic properties, including absorption, distribution, metabolism, and excretion.

Absorption. Several of the antifungal agents, including the polyene and echinocandin classes, do not have appreciable oral bioavailability. Until the early 1990s, the lack of oral treatment options left intravenous therapy as the only alternative for the treatment of invasive fungal infections. Today, each member of the azole class can be administered orally; however, the degree of absorption and optimal administration conditions vary for each of these drugs (table 2). Differences can even exist between various formulations of the same agent.

Table 2

Comparative pharmacokinetics of the antifungal agents.

Fluconazole is readily absorbed, with oral bioavailability easily achieving concentrations equal to 90% of those achieved by intravenous administration [37]. Absorption is not affected by food consumption, gastric pH, or disease state [38, 39]. Variable gastrointestinal absorption does occur with the other members of this class, however, and, for one compound (itraconazole), it varies according to the specific formulation. Oral bioavailability of these agents can be also be affected by food consumption and changes in gastric pH.

Itraconazole capsules demonstrate optimal absorption in the presence of gastric acid and, therefore, cannot be coadministered with agents known to raise gastric pH, such as H2 receptor antagonists or proton pump inhibitors [72, 73]. Furthermore, itraconazole capsules should be administered after a full meal to optimize absorption [74]. In general, the cyclodextrin solution is more efficiently absorbed (i.e., the area under the concentration curve [AUC] is increased by 30%) than is the capsule formulation [40]. In addition, antacid therapy does not have a negative effect on absorption [41, 75]. Food can decrease serum concentrations of itraconazole solution; therefore, this preparation should be administered on an empty stomach [42, 43].

The oral bioavailability of voriconazole is >90% when the stomach is empty, but it decreases when food is present [44, 45]. Thus, this agent should be administered on an empty stomach. In contrast, posaconazole absorption is optimized when administered with a high-fat meal or a similar composition nutritional supplement, such as Boost Plus (Novartis Nutrition) [46].

Distribution. The distribution of antifungal agents in the body is another important factor to consider in the treatment of invasive fungal infections, because these infections may occur at physiologically sequestered sites. As demonstrated by relatively large volumes of distribution, the available antifungal agents are widely distributed throughout the body, with a few significant exceptions discussed below [37, 41, 45, 47, 48]. The main factors affecting drug distribution are molecular size, charge, degree of protein binding, and route of elimination.

Fungal infections of the CNS are associated with high morbidity and mortality and are difficult to treat. Many antifungal agents have large molecular weights that preclude their ability to penetrate the blood-brain barrier and achieve therapeutic CSF concentrations. Currently, flucytosine, fluconazole, and voriconazole have the best CSF penetration, with each resulting in concentrations of at least 50% of those seen in serum [49, 76, 77]. The concept of CSF concentrations predicting the efficacy of antifungal agents for CNS infections is a bit misleading. For example, amphotericin B, a drug that is essentially undetectable in CSF, has been the mainstay of treatment for cryptococcal meningitis, despite the lack of detectable drug concentrations in the CSF [31, 78]. In these instances, it is postulated that tissue concentrations of these agents are adequate to allow efficacy. Recent investigations have suggested that brain parenchymal concentrations of the azole agents and echinocandins may be more meaningful for prediction of therapeutic response.

Ophthalmologic fungal infections are also difficult to treat. Traditionally, topical therapies have been used for these infections, especially when the disease is limited to superficial infection. Many of the available systemic antifungal therapies can achieve intraocular concentrations adequate for treatment of more invasive disease (table 2). For other agents, localized therapy, such as intravitreal injections, is required for reliable concentrations within the vitreous body.

Relatively few available antifungal agents are renally eliminated as unchanged drug or active metabolite and, therefore, do not provide high concentrations of microbiologically active drug in the urine (table 2). Currently, fluconazole and flucytosine are the only drugs that can achieve reliable urine concentrations >50% of serum exposure when given systemically [48, 50]. It is important to note that, because many of these agents produce adequate tissue concentrations, a lack of detectable urine concentrations does not necessarily preclude use when the disease involves renal parenchyma.

The degree of protein binding is another characteristic that alters systemic exposure to drug; a protein-bound drug is not available for microbiologic activity. Thus, this factor plays an important role in determining the amount of active drug present at a given site of infection [79]. Unfortunately, the majority of available pharmacokinetic data for the antifungal agents reflect concentrations of total drug. Therefore, clinicians are left to hypothesize about the amount of measured drug actually available to fight infection (i.e., the portion of free, unbound drug). The polyene agents and many azole antifungals, with the exception of voriconazole and fluconazole, are highly protein bound (>90%). Protein binding with the echinocandin class varies from 85% to 99% for anidulafungin, caspofungin, and micafungin [4, 46, 47, 51–53, 80, 81].

The major protein that binds these drugs is albumin, although other serum proteins may also play a role [82, 83]. Many patients who are at risk for fungal infection are malnourished and, as a result, have low levels of serum albumin. The effect of this on protein binding of drugs may result in higher concentrations of available or active drug; however, this concept has not been sufficiently studied with regard to a potential effect on antifungal drug dosing or efficacy [79, 84–86].

Metabolism and elimination. Many systemic antifungal agents undergo some degree of hepatic metabolism before elimination. One notable exception is flucytosine, which is not known to be metabolized hepatically, because urine excretion of unchanged drug accounts for >90% of its elimination [48]. For the amphotericin B products, the exact routes of metabolism and elimination are largely unknown [4].

All azole antifungals undergo some degree of hepatic metabolism (table 2). For fluconazole, the role of metabolism in drug elimination is minimal, but this is not the case with itraconazole, voriconazole, and posaconazole, which are highly dependent on metabolism for drug elimination. Given that there are few active antifungal metabolites, this results in production of inactive compounds that provide no clinically meaningful activity, with the notable exception of hydroxyitraconazole (a metabolite of itraconazole) [87]. Although oxidative metabolism is the primary process involved in azole metabolism, glucuronide conjugation does occur with some of these drugs, especially posaconazole [88].

Each of the 2 available echinocandins (caspofungin and micafungin) undergoes metabolism to produce 2 distinct inactive metabolites. For caspofungin, these processes are hepatic hydrolysis and N-acetylation [89]. Micafungin undergoes nonoxidative metabolism to produce 2 distinct compounds [90]. Although it is a weak substrate for cytochrome P450 (CYP450), the metabolism of micafungin does not appear to be affected by inhibitors or substrates of this enzyme system. Unlike caspofungin and micafungin, anidulafungin is not hepatically metabolized but undergoes nonenzymatic degradation [91].

Effect of organ dysfunction on drug dosing. The effect of organ dysfunction on the elimination of the antifungal agents is summarized in table 3. Although the various formulations of amphotericin B are known for their ability to cause nephrotoxicity, they do not require dose adjustment for patients with decreased renal function. In fact, of all the available systemic antifungal agents, only fluconazole and flucytosine require dosing modification when given to patients with decreased levels of creatinine clearance [4, 48]. In some instances, such as with amphotericin B, dosing regimens may be altered in attempts to ameliorate toxicity, but this is not done as a result of altered drug clearance. Another example is the cyclodextrins, which are present in the intravenous preparations of itraconazole and voriconazole and can accumulate in renal disease. Therefore, the use of these formulations in patients with creatinine clearance <50 mL/min, in the case of voriconazole, and 30 mL/min, in the case of itraconazole, is cautioned for these formulations [41, 45].

Table 3

Suggested dose modifications for antifungal agents, by type of organ dysfunction.

Hepatic disease can also affect the elimination of several antifungal agents. For the majority of these agents, however, no dose alteration is recommended. Of the azoles, only voriconazole requires dose reduction for patients with mild-to-moderate cirrhosis [45]. Similarly, caspofungin is the only echinocandin with recommendations for dose modification in severe hepatic disease [47]. The metric used to determine appropriate dosing in hepatic disease is the Child-Pugh scoring system, which is appropriate for patients with chronic liver dysfunction but not for patients who have acute hepatic injury. Currently, information is not available to guide drug dosing in this clinical scenario.

Drug-drug interactions. The effect of antifungal agents on other therapeutic regimens merits serious consideration when therapy is being initiated or discontinued. Antifungal drugs can alter the safety or efficacy of concomitant therapies through several mechanisms. The first of these involves additive toxicities associated with concomitant administration; the most apparent example is nephrotoxicity caused by amphotericin B. This toxicity can enhance the renal effect of many other agents, including cyclosporine and the aminoglycosides [93].

A more complicated issue relates to the inhibition of drug metabolism that occurs as a result of these drug interactions. A complete review of CYP450-mediated drug interactions is beyond the scope of this article, but the importance of this effect should not be minimized [94]. Because of their mechanism of action, all the azole antifungals inhibit CYP450 enzymes to some degree (table 4). As a result, careful consideration must be given when an azole agent is added to a patient's drug regimen. Similarly, when an azole agent is discontinued, the change in metabolism that occurs may have profound clinical implications. For example, organ rejection has been reported after discontinuation of an azole antifungal that was not accompanied by the necessary upward dose adjustments in the affected immunosuppressant agent (e.g., calcineurin inhibitor) [97].

Table 4

Summary of azole-mediated cytochrome P450 drug-drug interactions.

Although caspofungin and micafungin are not major substrates for the CYP450 enzyme system, they both have interactions that appear to be mediated via this mechanism. Caspofungin concentrations are decreased when administered with CYP450 inducers, such as rifampin and phenytoin [98]. Micafungin does have weak inhibitory properties against CYP3A4 and has been shown to increase serum concentrations of substrates of this enzyme. This phenomenon has been specifically assessed with sirolimus and nifedipine, and, in both cases, the AUC of the target drug was significantly elevated [54]. Anidulafungin does not appear to exhibit these CYP450-mediated interactions [91].

Initial product labeling for caspofungin indicated that a drug-drug interaction occurred when this agent was administered in combination with cyclosporine. This was based on data obtained in studies of healthy volunteers who received these drugs in combination as part of phase 1 development of caspofungin. After coadministration, an unacceptable elevation in hepatic enzymes was seen; therefore, cyclosporine was prohibited in clinical trials of caspofungin. This prohibition was reflected in the warning section of the initial caspofungin package insert [47]. More recent data, however, suggest that this effect is not reproducible in infected patients receiving the drug [99, 100]. In consideration of these data, these warnings have recently been revised in the product labeling.

Another mechanism involved in drug-drug interactions relates to the role of P-glycoprotein (P-gp). P-gp is a transporter protein involved in the absorption and distribution of drugs, as well as drug resistance. In a fashion similar to their interactions via the CYP450 enzyme system, azole antifungals have affinity for P-gp, because of their mechanism of action at the fungal cell membrane. More specifically, itraconazole is both a substrate and an inhibitor of P-gp, whereas fluconazole does not inhibit P-gp but may be a weak substrate. Therefore, interactions among the azole antifungals with P-gp may affect response to therapy or may play a role in interactions with other medications [101].

Pharmacodynamics. Another important consideration in the optimization of antifungal treatment regimens is the interaction between the fungal pathogen, the antifungal agent, and host factors. These pharmacodynamic principles have not been described for antifungal agents with the same level of detail as for the antibacterial agents. However, fairly extensive in vitro and animal model investigations have been undertaken with agents from the triazole, polyene, and echinocandin antifungal classes.

A series of reports has defined the pharmacokinetic exposure of these compounds relative to the MIC of the infecting pathogen as a means of optimizing treatment efficacy. In animal models of disseminated candidiasis, killing of fungal organisms with echinocandins and polyenes is optimized by achieving peak drug concentrations 2–10-fold in excess of the MIC [102, 103]. Treatment outcome with the triazole antifungals has been shown to correlate with the drug exposure over time, which is similar to the concentration needed to inhibit the organism in vitro, or the MIC. The pharmacokinetic index that best accounts for the entire exposure over time is the ratio of the 24-h AUC to the MIC (24-h AUC : MIC). In preclinical infection models, a free drug 24-h AUC : MIC value near 25 : 1 has been shown to reproducibly predict outcome with each of the triazole compounds [104]. Examination of clinical trial data with Candida infections has suggested that this pharmacodynamic relationship is similarly helpful for prediction of treatment efficacy in humans [105, 106]. The clinical relevance of the relationships between a specific drug exposure, the MIC, and outcome is less clear for other fungal pathogens and drug classes.

The relationship between antifungal pharmacokinetics and certain host toxicities has been demonstrated for a few compounds. Several decades ago, the relationship between flucytosine serum concentrations and bone marrow toxicity was elucidated [107]. More recently, the toxicodynamic relationship between higher-than-anticipated voriconazole exposure and hepatoxicity has been suggested [108]. More extensive evaluation of clinical data will be necessary to better understand these important pharmacodynamic relationships.

To aid clinicians in implementing these principles, serum drug concentration monitoring is now available for several of the available antifungal agents. Table 5 outlines the appropriate conditions for monitoring, target concentrations, and the association between this information and clinical outcomes, either therapeutic or toxic.

Table 5

Serum drug concentration monitoring for antifungal agents.

Toxicities

Table 6 reviews the major comparative toxicities of the systemic antifungal agents available for management of invasive fungal disease. In addition to the types of toxicities presented (i.e., hepatic, renal, hematologic, and infusional toxicities and electrolyte abnormalities), each agent is associated with a set of unique adverse events, as described below.

Table 6

Comparative toxicities of antifungal agents.

Amphotericin B preparations. The toxicity of amphotericin B is well known. In addition to the nephrotoxicity and acute infusion-related reactions associated with the drug, a unique pulmonary reaction can be seen, particularly with certain lipid preparations. With the liposomal preparation of amphotericin B, a triad of infusional toxicity has been characterized. This toxicity can manifest as a combination of the following clinical scenarios: pulmonary toxicity (i.e., chest pain, dyspnea, and hypoxia); abdominal, flank, or leg pain; or flushing and urticaria [119, 120]. Similarly, with amphotericin B colloidal dispersion, severe hypoxia has been reported in patients; in one study, hypoxia occurred more commonly in association with the use of amphotericin B colloidal dispersion than with amphotericin B deoxycholate [121]. Hypoxia has also been reported in association with use of the lipid complex of amphotericin B. In one study, up to 20% of patients experienced this toxicity. Unique characteristics in this case included onset of symptoms beyond the second day of therapy for >70% of patients [111].

Azole antifungal agents. Fluconazole is an extremely well-tolerated agent that lacks significant toxicity, despite having been used for treatment and prophylaxis in many patient populations for more than a decade. However, reversible alopecia is not uncommon with this agent [122].

Oral itraconazole solution is also relatively safe but can be associated with nausea and diarrhea severe enough to force discontinuation. This reaction is caused by the excipient hydroxypropyl-β-cyclodextrin, which is used to increase solubility of the parent drug [123]. Itraconazole has been described as causing a unique triad of hypertension, hypokalemia, and edema, mostly in older adults [124]. A negative inotropic effect resulting in congestive heart failure has also been described and has prompted changes to the package labeling to avoid administration of itraconazole to patients with a history of heart failure [41, 125].

Two unique adverse events have been associated with the use of voriconazole: visual disturbances and cutaneous phototoxicity. The mechanism for visual disturbances is not known but manifests itself as photopsia (i.e., the appearance of bright lights, color changes, or wavy lines) or abnormal vision in up to 45% of patients receiving the treatment [126]. This effect is usually mild and transient, and it abates with continued treatment. In addition, this effect appears to be associated with higher doses of voriconazole [112]. Rash has been reported in association with voriconazole use in up to 8% of subjects; phototoxicity-related rash occurs less frequently but is a significant problem for ambulatory patients [127, 128]. This effect is not prevented through the use of sunscreens but is reversible after discontinuation of therapy.

Posaconazole has been well tolerated in clinical trials to date. The most frequently reported adverse events attributed to the drug have been associated with hepatic toxicities. These toxicities seem to occur less frequently than with other members of the triazole class [113]. Fatal hepatotoxicity has been reported with itraconazole, voriconazole, and posaconazole. Therefore, close monitoring of hepatic function is warranted with all members of the azole class [41, 45].

Echinocandins. The echinocandins are associated with few toxicities, making them safe agents to administer. The most notable, yet uncommon, event reported is a histamine-mediated infusion-related reaction. As with vancomycin, this reaction can be relieved by slowing the rate of infusion or premedicating with an antihistamine, such as diphenhydramine.

Conclusion

Clinicians now have access to an expanded number of antifungal agents; however, the panacea of antifungal therapy remains to be found. Therefore, a keen appreciation of the properties associated with each antifungal agent is imperative in the selection and administration of antifungal therapy. Differences in the pharmacokinetics of each unique drug render effective administration a challenge, particularly given the complex regimens that patients who are at risk for fungal infection receive because of their underlying disease states. Toxicity profiles also play a major role in the treatment of fungal disease, and differences among the antifungal classes, as well as agents within a given class, must be understood. With judicious use of the available agents, we are able to successfully and safely treat a growing number of life-threatening infections.

Acknowledgments

Financial support. This article was supported by an educational grant from Schering-Plough.

Potential conflicts of interest. E.S.D.A. has received grants, research support, consultation fees, and honoraria from Pfizer, Astellas Pharma US, Enzon Pharmaceuticals, Merck, and Schering-Plough and is on the paid speakers' bureaus of Pfizer, Astellas Pharma US, Enzon Pharmaceuticals, and Schering-Plough. R.L. has received grants and research support from Merck, Pfizer, Astellas Pharma US, and Enzon Pharmaceuticals; has received consultation fees from Merck, Schering-Plough, Pfizer, and Enzon Pharmaceuticals; has received honoraria from Merck, Pfizer, and Schering-Plough; and is on the paid speakers' bureaus of Merck and Astellas Pharma US. J.S.L. has received honoraria and consultation fees from Pfizer, Astellas Pharma US, and Schering-Plough and is on the paid speakers' bureaus of Pfizer, Astellas Pharma US, and Schering-Plough. C.M. has received grants and research support from Ortho-McNeil; has received consultation fees from Wyeth Pharmaceuticals, Ortho-McNeil, and Elan Pharmaceuticals; has received honoraria from Pfizer and Ortho-McNeil; and is on the paid speakers' bureaus of Ortho-McNeil, Wyeth Pharmaceuticals, and Elan Pharmaceuticals. D.A. has received grants and research support from Vicuron Pharmaceuticals, Schering-Plough, Pfizer, GlaxoSmithKline, Astellas Pharma US, and Enzon Pharmaceuticals; has received consultation fees from Vicuron Pharmaceuticals, Merck, Schering-Plough, Bristol-Myers Squibb Cytogenetics, Conjugon, Pfizer, and Astellas Pharma US; and has received honoraria from Merck and Pfizer.

References

1
Center for Drug Evaluation and Research
New and generic drug approvals: 1998–2004
2004
Washington, DC
US Food and Drug Administration
 
Available at: http://www.fda.gov/cder/approval/index.htm. Accessed 23 February 2006
2
Hoffman
JM
Shah
ND
Vermeulen
LC
Hunkler
RJ
Hontz
KM
Projecting future drug expenditures—2005
Am J Health Syst Pharm
2005
, vol. 
62
 (pg. 
149
-
67
)
3
Gallis
HA
Drew
RH
Pickard
WW
Amphotericin B: 30 years of clinical experience
Rev Infect Dis
1990
, vol. 
12
 (pg. 
308
-
29
)
4
Groll
AH
Piscitelli
SC
Walsh
TJ
Clinical pharmacology of systemic antifungal agents: a comprehensive review of agents in clinical use, current investigational compounds, and putative targets for antifungal drug development
Adv Pharmacol
1998
, vol. 
44
 (pg. 
343
-
500
)
5
Vermes
A
Guchelaar
H-J
Dankert
J
Flucytosine: a review of its pharmacology, clinical indications, pharmacokinetics, toxicity and drug interactions
J Antimicrob Chemother
2000
, vol. 
46
 (pg. 
171
-
9
)
6
Pfaller
MA
Messer
SA
Hollis
RJ
Jones
RN
In vitro activities of posaconazole (Sch 56592) compared with those of itraconazole and fluconazole against 3,685 clinical isolates of Candida spp. and Cryptococcus neoformans
Antimicrob Agents Chemother
2001
, vol. 
45
 (pg. 
2862
-
4
)
7
Pfaller
MA
Messer
SA
Hollis
RJ
Jones
RN
Antifungal activities of posaconazole, ravuconazole, and voriconazole compared to those of itraconazole and amphotericin B against 239 clinical isolates of Aspergillus spp. and other filamentous fungi: report from SENTRY Antimicrobial Surveillance Program, 2000
Antimicrob Agents Chemother
2002
, vol. 
46
 (pg. 
1032
-
7
)
8
Arikan
S
Paetznick
VL
Rex
JH
Comparative evaluation of disk diffusion with microdilution assay in susceptibility testing of caspofungin against Aspergillus and Fusarium isolates
Antimicrob Agents Chemother
2002
, vol. 
46
 (pg. 
3084
-
7
)
9
Pfaller
MA
Jones
RN
Doern
GV
Sader
HS
Hollis
RJ
Messer
SA
International surveillance of bloodstream infections due to Candida species: frequency of occurrence and antifungal susceptibilities of isolates collected in 1997 in the United States, Canada, and South America for the SENTRY Program. SENTRY Participant Group
J Clin Microbiol
1998
, vol. 
36
 (pg. 
1886
-
9
)
10
Pfaller
MA
Jones
RN
Doern
GV
, et al. 
International surveillance of blood stream infections due to Candida species in the European SENTRY program: species distribution and antifungal susceptibility including the investigational triazole and echinocandin agents. SENTRY Participant Group (Europe)
Diagn Microbiol Infect Dis
1999
, vol. 
35
 (pg. 
19
-
25
)
11
van Duin
D
Cleare
W
Zaragoza
O
Casadevall
A
Nosanchuk
JD
Effects of voriconazole on Cryptococcus neoformans
Antimicrob Agents Chemother
2004
, vol. 
48
 (pg. 
2014
-
20
)
12
Proia
LA
Tenorio
AR
Successful use of voriconazole for treatment of Coccidioides meningitis
Antimicrob Agents Chemother
2004
, vol. 
48
 pg. 
2341
 
13
Lass-Flörl
C
Nagl
M
Speth
C
Ulmer
H
Dierich
M
Würzner
R
Studies of in vitro activities of voriconazole and itraconazole against Aspergillus hyphae using viability staining
Antimicrob Agents Chemother
2001
, vol. 
45
 (pg. 
124
-
8
)
14
González
GM
Fothergill
AW
Sutton
DA
Rinaldi
MG
Loebenberg
D
In vitro activities of new and established triazoles against opportunistic filamentous and dimorphic fungi
Med Mycol
2005
, vol. 
43
 (pg. 
281
-
4
)
15
Pfaller
MA
Messer
SA
Boyken
L
Huynh
H
Hollis
RJ
Diekema
DJ
In vitro activities of 5-fluorocytosine against 8,803 clinical isolates of Candida spp.: global assessment of primary resistance using National Committee for Clinical Laboratory Standards susceptibility testing methods
Antimicrob Agents Chemother
2002
, vol. 
46
 (pg. 
3518
-
21
)
16
Espinel-Ingroff
A
Comparison of in vitro activities of the new triazole SCH56592 and the echinocandins MK-0991 (L-743,872) and LY303366 against opportunistic filamentous and dimorphic fungi and yeasts
J Clin Microbiol
1998
, vol. 
36
 (pg. 
2950
-
6
)
17
Tawara
S
Ikeda
F
Maki
K
, et al. 
In vitro activities of a new lipopeptide antifungal agent, FK463, against a variety of clinically important fungi
Antimicrob Agents Chemother
2000
, vol. 
44
 (pg. 
57
-
62
)
18
Rex
JH
Pfaller
MA
Barry
AL
Nelson
PW
Webb
CD
Antifungal susceptibility testing of isolates from a randomized, multicenter trial of fluconazole versus amphotericin B as treatment of nonneutropenic patients with candidemia. NIAID Mycoses Study Group and the Candidemia Study Group
Antimicrob Agents Chemother
1995
, vol. 
39
 (pg. 
40
-
4
)
19
Pfaller
MA
Bale
MJ
Buschelman
B
Rhomberg
P
Antifungal activity of a new triazole, D0870, compared with four other antifungal agents tested against clinical isolates of Candida and Torulopsis glabrata
Diagn Microbiol Infect Dis
1994
, vol. 
19
 (pg. 
75
-
80
)
20
Martinez-Suarez
JV
Rodriguez-Tudela
JL
Patterns of in vitro activity of itraconazole and imidazole antifungal agents against Candida albicans with decreased susceptibility to fluconazole from Spain
Antimicrob Agents Chemother
1995
, vol. 
39
 (pg. 
1512
-
6
)
21
Pappas
PG
Rex
JH
Sobel
JD
, et al. 
Guidelines for treatment of candidiasis
Clin Infect Dis
2004
, vol. 
38
 (pg. 
161
-
89
)
22
Ostrosky-Zeichner
L
Rex
JH
Pappas
PG
, et al. 
Antifungal susceptibility survey of 2,000 bloodstream Candida isolates in the United States
Antimicrob Agents Chemother
2003
, vol. 
47
 (pg. 
3149
-
54
)
23
Nakai
T
Uno
J
Otomo
K
, et al. 
In vitro activity of FK463, a novel lipopeptide antifungal agent, against a variety of clinically important molds
Chemotherapy
2002
, vol. 
48
 (pg. 
78
-
81
)
24
Rex
JH
Cooper
CR
Jr
Merz
WG
Galgiani
JN
Anaissie
EJ
Detection of amphotericin B-resistant Candida isolates in a broth-based system
Antimicrob Agents Chemother
1995
, vol. 
39
 (pg. 
906
-
9
)
25
González
GM
Tijerina
R
Najvar
LK
, et al. 
Correlation between antifungal susceptibilities of Coccidioides immitis in vitro and antifungal treatment with caspofungin in a mouse model
Antimicrob Agents Chemother
2001
, vol. 
45
 (pg. 
1854
-
9
)
26
Antony
S
Use of the echinocandins (caspofungin) in the treatment of disseminated coccidioidomycosis in a renal transplant recipient
Clin Infect Dis
2004
, vol. 
39
 (pg. 
879
-
80
)
27
Ng
AWK
Wasan
KM
Lopez-Berestein
G
Development of liposomal polyene antibiotics: an historical perspective
J Pharm Pharm Sci
2003
, vol. 
6
 (pg. 
67
-
83
)
28
Deray
G
Amphotericin B nephrotoxicity
J Antimicrob Chemother
2002
, vol. 
49
 
Suppl S1
(pg. 
37
-
41
)
29
Johnson
PC
Wheat
LJ
Cloud
GA
, et al. 
Safety and efficacy of liposomal amphotericin B compared with conventional amphotericin B for induction therapy of histoplasmosis in patients with AIDS
Ann Intern Med
2002
, vol. 
137
 (pg. 
1
-
105
)
30
Waldorf
AR
Polak
A
Mechanisms of action of 5-fluorocytosine
Antimicrob Agents Chemother
1983
, vol. 
23
 (pg. 
79
-
85
)
31
Saag
MS
Graybill
RJ
Larsen
RA
, et al. 
Practice guidelines for the management of cryptococcal disease. Mycoses Study Group Cryptococcal Subproject of the National Institute of Allergy and Infectious Diseases
Clin Infect Dis
2000
, vol. 
30
 (pg. 
710
-
8
)
32
Tassel
D
Madoff
MA
Treatment of Candida sepsis and Cryptococcus meningitis with 5-fluorocytosine: a new antifungal agent
JAMA
1968
, vol. 
206
 (pg. 
830
-
2
)
33
Polak
A
Scholer
HJ
Mode of action of 5-fluorocytosine and mechanisms of resistance
Chemotherapy
1975
, vol. 
21
 (pg. 
113
-
30
)
34
Sheehan
DJ
Hitchcock
CA
Sibley
CM
Current and emerging azole antifungal agents
Clin Microbiol Rev
1999
, vol. 
12
 (pg. 
40
-
79
)
35
Manavathu
EK
Cutright
JL
Chandrasekar
PH
Organism-dependent fungicidal activities of azoles
Antimicrob Agents Chemother
1998
, vol. 
42
 (pg. 
3018
-
21
)
36
Barchiesi
F
Schimizzi
AM
Fothergill
AW
Scalise
G
Rinaldi
MG
In vitro activity of the new echinocandin antifungal, MK-0991, against common and uncommon clinical isolates of Candida species
Eur J Clin Microbiol Infect Dis
1999
, vol. 
18
 (pg. 
302
-
4
)
37
Diflucan [package insert]
2004
New York, NY
Roerig
38
Zimmermann
T
Yeates
RA
Laufen
H
Pfaff
G
Wildfeuer
A
Influence of concomitant food intake on the oral absorption of two triazole antifungal agents, itraconazole and fluconazole
Eur J Clin Pharmacol
1994
, vol. 
46
 (pg. 
147
-
50
)
39
Zimmermann
T
Yeates
RA
Riedel
KD
Lach
P
Laufen
H
The influence of gastric pH on the pharmacokinetics of fluconazole: the effect of omeprazole
Int J Clin Pharmacol Ther
1994
, vol. 
32
 (pg. 
491
-
6
)
40
Barone
JA
Moskovitz
BL
Guarnieri
J
, et al. 
Enhanced bioavailability of itraconazole in hydroxypropyl-β-cyclodextrin solution versus capsules in healthy volunteers
Antimicrob Agents Chemother
1998
, vol. 
42
 (pg. 
1862
-
5
)
41
Sporanox [package insert]
2004
Titusville, NJ
Janssen Pharmaceutica
42
Barone
JA
Moskovitz
BL
Guarnieri
J
, et al. 
Food interaction and steady-state pharmacokinetics of itraconazole oral solution in healthy volunteers
Pharmacotherapy
1998
, vol. 
18
 (pg. 
295
-
301
)
43
Van de Velde
VJ
Van Peer
AP
Heykants
JJ
, et al. 
Effect of food on the pharmacokinetics of a new hydroxypropyl-β-cyclodextrin formulation of itraconazole
Pharmacotherapy
1996
, vol. 
16
 (pg. 
424
-
8
)
44
Purkins
L
Wood
N
Kleinermans
D
Greenhalgh
K
Nichols
D
Effect of food on the pharmacokinetics of multiple-dose oral voriconazole
Br J Clin Pharmacol
2003
, vol. 
56
 (pg. 
17
-
23
)
45
Vfend [package insert]
2004
New York, NY
Roeirg
46
Courtney
R
Wexler
D
Radwanski
E
Lim
J
Laughlin
M
Effect of food on the relative bioavailability of two oral formulations of posaconazole in healthy adults
Br J Clin Pharmacol
2004
, vol. 
57
 (pg. 
218
-
22
)
47
Cancidas [package insert]
2005
Whitehouse Station, NJ
Merck
48
Ancobon [package insert]
2003
Costa Mesa, CA
ICN Pharmaceuticals
49
Lutsar
I
Roffey
S
Troke
P
Voriconazole concentrations in the cerebrospinal fluid and brain tissue of guinea pigs and immunocompromised patients
Clin Infect Dis
2003
, vol. 
37
 (pg. 
728
-
32
)
50
Brammer
KW
Coakley
AJ
Jezequel
SG
Tarbit
MH
The disposition and metabolism of [14C]fluconazole in humans
Drug Metab Dispos
1991
, vol. 
19
 (pg. 
764
-
7
)
51
Amphocin [package insert]
2003
Kalamazoo, MI
Pharmacia & Upjohn
52
Abelcet [package insert]
2002
Piscataway, NJ
Enzon
53
Amphotec [package insert]
2001
Brisbane, CA
InterMune
54
Mycamine [package insert]
2005
Deerfield, IL
Astellas Pharma US
55
Groll
AH
Mickiene
D
Petraitiene
R
, et al. 
Pharmacokinetic and pharmacodynamic modeling of anidulafungin (LY303366): reappraisal of its efficacy in neutropenic animal models of opportunistic mycoses using optimal plasma sampling
Antimicrob Agents Chemother
2001
, vol. 
45
 (pg. 
2845
-
55
)
56
Groll
AH
Mickiene
D
Petraitis
V
, et al. 
Compartmental pharmacokinetics and tissue distribution of the antifungal echinocandin lipopeptide micafungin (FK463) in rabbits
Antimicrob Agents Chemother
2001
, vol. 
45
 (pg. 
3322
-
7
)
57
Bekersky
I
Fielding
RM
Dressler
DE
Lee
JW
Buell
DN
Walsh
TJ
Pharmacokinetics, excretion, and mass balance of liposomal amphotericin B (AmBisome) and amphotericin B deoxycholate in humans
Antimicrob Agents Chemother
2002
, vol. 
46
 (pg. 
828
-
33
)
58
AmBisome [package insert]
2005
San Dimas, CA
Gilead Sciences
59
Groll
AH
Giri
N
Gonzalez
CE
, et al. 
Penetration of lipid formulations of amphotericin B into cerebrospinal fluid and brain tissue [abstract A90]
Program and abstracts of the 37th Interscience Conference on Antimicrobial Agents and Chemotherapy (Toronto)
1997
Washington, DC
American Society for Microbiology
60
Goldblum
D
Rohrer
K
Frueh
BE
Theurillat
R
Thormann
W
Zimmerli
S
Ocular distribution of intravenously administered lipid formulations of amphotericin B in a rabbit model
Antimicrob Agents Chemother
2002
, vol. 
46
 (pg. 
3719
-
23
)
61
Louie
A
Liu
W
Miller
DA
, et al. 
Efficacies of high-dose fluconazole plus amphotericin B and high-dose fluconazole plus 5-fluorocytosine versus amphotericin B, fluconazole, and 5-fluorocytosine monotherapies in treatment of experimental endocarditis, endophthalmitis, and pyelonephritis due to Candida albicans
Antimicrob Agents Chemother
1999
, vol. 
43
 (pg. 
2831
-
40
)
62
O'Day
DM
Foulds
G
Williams
TE
Robinson
RD
Allen
RH
Head
WS
Ocular uptake of fluconazole following oral administration
Arch Ophthalmol
1990
, vol. 
108
 (pg. 
1006
-
8
)
63
Fisher
JF
Taylor
AT
Clark
J
Rao
R
Espinel-Ingroff
A
Penetration of amphotericin B into the human eye
J Infect Dis
1983
, vol. 
147
 pg. 
164
 
64
Walsh
A
Haft
DA
Miller
MH
Loran
MR
Friedman
AH
Ocular penetration of 5-fluorocytosine
Invest Ophthalmol Vis Sci
1978
, vol. 
17
 (pg. 
691
-
4
)
65
Hariprasad
SM
Mieler
WF
Holz
ER
, et al. 
Determination of vitreous, aqueous, and plasma concentration of orally administered voriconazole in humans
Arch Ophthalmol
2004
, vol. 
122
 (pg. 
42
-
7
)
66
Savani
DV
Perfect
JR
Cobo
LM
Durack
DT
Penetration of new azole compounds into the eye and efficacy in experimental Candida endophthalmitis
Antimicrob Agents Chemother
1987
, vol. 
31
 (pg. 
6
-
10
)
67
Mian
UK
Mayers
M
Garg
Y
, et al. 
Comparison of fluconazole pharmacokinetics in serum, aqueous humor, vitreous humor, and cerebrospinal fluid following a single dose and at steady state
J Ocul Pharmacol Ther
1998
, vol. 
14
 (pg. 
459
-
71
)
68
Dowell
JA
Knebel
W
Ludden
T
Stogniew
M
Krause
D
Henkel
T
Population pharmacokinetic analysis of anidulafungin, an echinocandin antifungal
J Clin Pharmacol
2004
, vol. 
44
 (pg. 
590
-
8
)
69
Cutler
RE
Blair
AD
Kelly
MR
Flucytosine kinetics in subjects with normal and impaired renal function
Clin Pharmacol Ther
1978
, vol. 
24
 (pg. 
333
-
42
)
70
Boucher
HW
Groll
AH
Chiou
CC
Walsh
TJ
Newer systemic antifungal agents: pharmacokinetics, safety and efficacy
Drugs
2004
, vol. 
64
 (pg. 
1997
-
2020
)
71
Gauthier
GM
Nork
TM
Prince
R
Andes
D
Subtherapeutic ocular penetration of caspofungin and associated treatment failure in Candida albicans endophthalmitis
Clin Infect Dis
2005
, vol. 
41
 (pg. 
e27
-
8
)
72
Jaruratanasirikul
S
Sriwiriyajan
S
Effect of omeprazole on the pharmacokinetics of itraconazole
Eur J Clin Pharmacol
1998
, vol. 
54
 (pg. 
159
-
61
)
73
Lange
D
Pavao
JH
Wu
J
Klausner
M
Effect of a cola beverage on the bioavailability of itraconazole in the presence of H2 blockers
J Clin Pharmacol
1997
, vol. 
37
 (pg. 
535
-
40
)
74
Van Peer
A
Woestenborghs
R
Heykants
J
Gasparini
R
Gauwenbergh
G
The effects of food and dose on the oral systemic availability of itraconazole in healthy subjects
Eur J Clin Pharmacol
1989
, vol. 
36
 (pg. 
423
-
6
)
75
Johnson
MD
Hamilton
CD
Drew
RH
Sanders
LL
Pennick
GJ
Perfect
JR
A randomized comparative study to determine the effect of omeprazole on the peak serum concentration of itraconazole oral solution
J Antimicrob Chemother
2003
, vol. 
51
 (pg. 
453
-
7
)
76
Arndt
CA
Walsh
TJ
McCully
CL
Balis
FM
Pizzo
PA
Poplack
DG
Fluconazole penetration into cerebrospinal fluid: implications for treating fungal infections of the central nervous system
J Infect Dis
1988
, vol. 
157
 (pg. 
178
-
80
)
77
Foulds
G
Brennan
DR
Wajszczuk
C
, et al. 
Fluconazole penetration into cerebrospinal fluid in humans
J Clin Pharmacol
1988
, vol. 
28
 (pg. 
363
-
6
)
78
Atkinson
AJ
Jr
Bennett
JE
Amphotericin B pharmacokinetics in humans
Antimicrob Agents Chemother
1978
, vol. 
13
 (pg. 
271
-
6
)
79
Wise
R
The clinical relevance of protein binding and tissue concentrations in antimicrobial therapy
Clin Pharmacokinet
1986
, vol. 
11
 (pg. 
470
-
82
)
80
Theuretzbacher
U
Pharmacokinetics/pharmacodynamics of echinocandins
Eur J Clin Microbiol Infect Dis
2004
, vol. 
23
 (pg. 
805
-
12
)
81
Murdoch
D
Plosker
GL
Anidulafungin
Drugs
2004
, vol. 
64
 (pg. 
2249
-
58
)
82
Bekersky
I
Fielding
RM
Dressler
DE
Lee
JW
Buell
DN
Walsh
TJ
Plasma protein binding of amphotericin B and pharmacokinetics of bound versus unbound amphotericin B after administration of intravenous liposomal amphotericin B (AmBisome) and amphotericin B deoxycholate
Antimicrob Agents Chemother
2002
, vol. 
46
 (pg. 
834
-
40
)
83
Hajdu
R
Thompson
R
Sundelof
JG
, et al. 
Preliminary animal pharmacokinetics of the parenteral antifungal agent MK-0991 (L-743,872)
Antimicrob Agents Chemother
1997
, vol. 
41
 (pg. 
2339
-
44
)
84
Schäfer-Korting
M
Korting
HC
Rittler
W
Obermuller
W
Influence of serum protein binding on the in vitro activity of anti-fungal agents
Infection
1995
, vol. 
23
 (pg. 
292
-
7
)
85
Schafer-Korting
M
Korting
HC
Amman
F
Peuser
R
Lukacs
A
Influence of albumin on itraconazole and ketoconazole antifungal activity: results of a dynamic in vitro study
Antimicrob Agents Chemother
1991
, vol. 
35
 (pg. 
2053
-
6
)
86
Zhanel
GG
Saunders
DG
Hoban
DJ
Karlowsky
JA
Influence of human serum on antifungal pharmacodynamics with Candida albicans
Antimicrob Agents Chemother
2001
, vol. 
45
 (pg. 
2018
-
22
)
87
Odds
FC
Vanden Bossche
H
Antifungal activity of itraconazole compared with hydroxy-itraconazole in vitro
J Antimicrob Chemother
2000
, vol. 
45
 (pg. 
371
-
3
)
88
Krieter
P
Flannery
B
Musick
T
Gohdes
M
Martinho
M
Courtney
R
Disposition of posaconazole following single-dose oral administration in healthy subjects
Antimicrob Agents Chemother
2004
, vol. 
48
 (pg. 
3543
-
51
)
89
Balani
SK
Xu
X
Arison
BH
, et al. 
Metabolites of caspofungin acetate, a potent antifungal agent, in human plasma and urine
Drug Metab Dispos
2000
, vol. 
28
 (pg. 
1274
-
8
)
90
Hebert
MF
Smith
HE
Marbury
TC
, et al. 
Pharmacokinetics of micafungin in healthy volunteers, volunteers with moderate liver disease, and volunteers with renal dysfunction
J Clin Pharmacol
2005
, vol. 
45
 (pg. 
1145
-
52
)
91
Stogniew
M
Pu
F
Henkel
T
Dowell
J
Anidulafungin biotransformation in humans is by degradation not metabolism [abstract P-1223]
Clin Microbiol Infect
2003
, vol. 
9
 
Suppl
pg. 
291
 
92
Courtney
R
Sansone
A
Smith
W
, et al. 
Posaconazole pharmacokinetics, safety, and tolerability in subjects with varying degrees of chronic renal disease
J Clin Pharmacol
2005
, vol. 
45
 (pg. 
185
-
92
)
93
Churchill
D
Seely
J
Nephrotoxicity associated with combined gentamicin-amphotericin B therapy
Nephron
1977
, vol. 
19
 (pg. 
176
-
81
)
94
Michalets
EL
Update: clinically significant cytochrome P-450 drug interactions
Pharmacotherapy
1998
, vol. 
18
 (pg. 
84
-
112
)
95
Wexler
D
Courtney
R
Richards
W
Banfield
C
Lim
J
Laughlin
M
Effect of posaconazole on cytochrome P450 enzymes: a randomized, open-label, two-way crossover study
Eur J Pharm Sci
2004
, vol. 
21
 (pg. 
645
-
53
)
96
Niwa
T
Shiraga
T
Takagi
A
Effect of antifungal drugs on cytochrome P450 (CYP) 2C9, CYP2C19, and CYP3A4 activities in human liver microsomes
Biol Pharm Bull
2005
, vol. 
28
 (pg. 
1805
-
8
)
97
Groll
AH
Kolve
H
Ehlert
K
Paulussen
M
Vormoor
J
Pharmacokinetic interaction between voriconazole and cyclosporin A following allogeneic bone marrow transplantation
J Antimicrob Chemother
2004
, vol. 
53
 (pg. 
113
-
4
)
98
Stone
JA
Migoya
EM
Hickey
L
, et al. 
Potential for interactions between caspofungin and nelfinavir or rifampin
Antimicrob Agents Chemother
2004
, vol. 
48
 (pg. 
4306
-
14
)
99
Anttila
VJ
Piilonen
A
Valtonen
M
Co-administration of caspofungin and cyclosporine to a kidney transplant patient with pulmonary Aspergillus infection
Scand J Infect Dis
2003
, vol. 
35
 (pg. 
893
-
4
)
100
Marr
KA
Hachem
R
Papanicolaou
G
, et al. 
Retrospective study of the hepatic safety profile of patients concomitantly treated with caspofungin and cyclosporin A
Transpl Infect Dis
2004
, vol. 
6
 (pg. 
110
-
6
)
101
Wang
EJ
Lew
K
Casciano
CN
Clement
RP
Johnson
WW
Interaction of common azole antifungals with P glycoprotein
Antimicrob Agents Chemother
2002
, vol. 
46
 (pg. 
160
-
5
)
102
Andes
D
Stamsted
T
Conklin
R
Pharmacodynamics of amphotericin B in a neutropenic-mouse disseminated-candidiasis model
Antimicrob Agents Chemother
2001
, vol. 
45
 (pg. 
922
-
6
)
103
Andes
D
Marchillo
K
Lowther
J
Bryskier
A
Stamstad
T
Conklin
R
In vivo pharmacodynamics of HMR 3270, a glucan synthase inhibitor, in a murine candidiasis model
Antimicrob Agents Chemother
2003
, vol. 
47
 (pg. 
1187
-
92
)
104
Andes
D
Marchillo
K
Conklin
R
, et al. 
Pharmacodynamics of a new triazole, posaconazole, in a murine model of disseminated candidiasis
Antimicrob Agents Chemother
2004
, vol. 
48
 (pg. 
137
-
42
)
105
Lee
S-C
Fung
C-P
Huang
J-S
, et al. 
Clinical correlates of antifungal macrodilution susceptibility test results for non-AIDS patients with severe Candida infections treated with fluconazole
Antimicrob Agents Chemother
2000
, vol. 
44
 (pg. 
2715
-
8
)
106
Rex
JH
Pfaller
M
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
)
107
Francis
P
Walsh
TJ
Evolving role of flucytosine in immunocompromised patients: new insights into safety, pharmacokinetics, and antifungal therapy
Clin Infect Dis
1992
, vol. 
15
 (pg. 
1003
-
18
)
108
Potoski
BA
Brown
J
The safety of voriconazole
Clin Infect Dis
2002
, vol. 
35
 (pg. 
1273
-
5
)
109
Andes
D
In vivo pharmacodynamics of antifungal drugs in treatment of candidiasis
Antimicrob Agents Chemother
2003
, vol. 
47
 (pg. 
1179
-
86
)
110
Denning
DW
Ribaud
P
Milpied
N
, et al. 
Efficacy and safety of voriconazole in the treatment of acute invasive aspergillosis
Clin Infect Dis
2002
, vol. 
34
 (pg. 
563
-
71
)
111
Wingard
JR
White
MH
Anaissie
E
Raffalli
J
Goodman
J
Arrieta
A
A randomized, double-blind comparative trial evaluating the safety of liposomal amphotericin B versus amphotericin B lipid complex in the empirical treatment of febrile neutropenia. L Amph/ABLC Collaborative Study Group
Clin Infect Dis
2000
, vol. 
31
 (pg. 
1155
-
63
)
112
Purkins
L
Wood
N
Ghahramani
P
Greenhalgh
K
Allen
MJ
Kleinermans
D
Pharmacokinetics and safety of voriconazole following intravenous- to oral-dose escalation regimens
Antimicrob Agents Chemother
2002
, vol. 
46
 (pg. 
2546
-
53
)
113
Graybill
JR
Raad
I
Negroni
R
Corcoran
G
Pedicone
L
Posaconazole (POS) long-term safety in subjects with invasive fungal infections (IFIs) [abstract M-1025]
Program and abstracts of the 44th Interscience Conference on Antimicrobial Agents and Chemotherapy (Washington, DC)
2004
Washington, DC
American Society for Microbiology
114
Tiphine
M
Letscher-Bru
V
Herbrecht
R
Amphotericin B and its new formulations: pharmacologic characteristics, clinical efficacy, and tolerability
Transpl Infect Dis
1999
, vol. 
1
 (pg. 
273
-
83
)
115
Ostrosky-Zeichner
L
Marr
KA
Rex
JH
Cohen
SH
Amphotericin B: time for a new “gold standard”
Clin Infect Dis
2003
, vol. 
37
 (pg. 
415
-
25
)
116
van Burik
JAH
Ratanatharathorn
V
Stepan
DE
, et al. 
Micafungin versus fluconazole for prophylaxis against invasive fungal infections during neutropenia in patients undergoing hematopoietic stem cell transplantation. National Institute of Allergy and Infectious Diseases Mycoses Study Group
Clin Infect Dis
2004
, vol. 
39
 (pg. 
1407
-
16
)
117
Wong-Beringer
A
Kriengkauykiat
J
Systemic antifungal therapy: new options, new challenges
Pharmacotherapy
2003
, vol. 
23
 (pg. 
1441
-
62
)
118
Krause
DS
Reinhardt
J
Vazquez
JA
, et al. 
Phase 2, randomized, dose-ranging study evaluating the safety and efficacy of anidulafungin in invasive candidiasis and candidemia
Antimicrob Agents Chemother
2004
, vol. 
48
 (pg. 
2021
-
4
)
119
White
MH
Bowden
RA
Sandler
ES
, et al. 
Randomized, double-blind clinical trial of amphotericin B colloidal dispersion vs. amphotericin B in the empirical treatment of fever and neutropenia
Clin Infect Dis
1998
, vol. 
27
 (pg. 
296
-
302
)
120
Johnson
MD
Drew
RH
Perfect
JR
Chest discomfort associated with liposomal amphotericin B: report of three cases and review of the literature
Pharmacotherapy
1998
, vol. 
18
 (pg. 
1053
-
61
)
121
Roden
MM
Nelson
LD
Knudsen
TA
, et al. 
Triad of acute infusion-related reactions associated with liposomal amphotericin B: analysis of clinical and epidemiological characteristics
Clin Infect Dis
2003
, vol. 
36
 (pg. 
1213
-
20
)
122
Pappas
PG
Kauffman
CA
Perfect
J
, et al. 
Alopecia associated with fluconazole therapy
Ann Intern Med
1995
, vol. 
123
 (pg. 
354
-
7
)
123
Vandewoude
K
Vogelaers
D
Decruyenaere
J
, et al. 
Concentrations in plasma and safety of 7 days of intravenous itraconazole followed by 2 weeks of oral itraconazole solution in patients in intensive care units
Antimicrob Agents Chemother
1997
, vol. 
41
 (pg. 
2714
-
8
)
124
Sharkey
PK
Rinaldi
MG
Dunn
JF
Hardin
TC
Fetchick
RJ
Graybill
JR
High-dose itraconazole in the treatment of severe mycoses
Antimicrob Agents Chemother
1991
, vol. 
35
 (pg. 
707
-
13
)
125
Ahmad
SR
Singer
SJ
Leissa
BG
Congestive heart failure associated with itraconazole
Lancet
2001
, vol. 
357
 (pg. 
1766
-
7
)
126
Herbrecht
R
Denning
DW
Patterson
TF
, et al. 
Voriconazole versus amphotericin B for primary therapy of invasive aspergillosis
N Engl J Med
2002
, vol. 
347
 (pg. 
408
-
15
)
127
Rubenstein
M
Levy
ML
Metry
D
Voriconazole-induced retinoid-like photosensitivity in children
Pediatr Dermatol
2004
, vol. 
21
 (pg. 
675
-
8
)
128
Denning
DW
Griffiths
CEM
Muco-cutaneous retinoid-effects and facial erythema related to the novel triazole antifungal agent voriconazole
Clin Exp Dermatol
2001
, vol. 
26
 (pg. 
648
-
53
)

Figures and Tables

Table 1

Antifungal spectrum of activity against common fungi.

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.