1,386
Views
24
CrossRef citations to date
0
Altmetric
Review

Direct effects of non-antifungal agents used in cancer chemotherapy and organ transplantation on the development and virulence of Candida and Aspergillus species

, &
Pages 280-295 | Received 15 Apr 2011, Accepted 03 Jun 2011, Published online: 01 Jul 2011

Abstract

Conventional antineoplastic, novel immunosuppressive agents and antibiotics used in cancer treatment can directly affect the growth, development and virulence of Candida and Aspergillus species. Cytotoxic and cisplatin compounds have anti-Candida activity and may be synergistic with antifungal drugs; they also inhibit Candida and Aspergillus filamentation/conidation and effect increased virulence in vitro. Glucocorticoids enhance Candida adherence to epithelial cells, germination in serum and in vitro secretion of phospholipases and proteases, as well as growth of A. fumigatus. Calcineurin and target of rapamycin inhibitors perturb Candida and Aspergillus morphogenesis, stress responses and survival in serum, reduce azole tolerance in Candida, but yield conflicting in vivo data. Inhibition of candidal heat shock protein 90 and candidal-specific histone deacetylase represent feasible therapeutic approaches for candidiasis. Tyrosine kinase inhibitors inhibit fungal cell entry into epithelial cells and phagocytosis. Quinolone and other antibiotics may augment activity of azole and polyene agents. The correlation of in vitro effects with clinically meaningful in vivo systems is warranted. 

Introduction

Patients receiving chemotherapy for cancer and individuals who are immunosuppressed after hematopoietic stem cell or organ transplantation are at increased risk for opportunistic fungal infections with high attendant morbidity and mortality.Citation1,Citation2 Although previously uncommon pathogens, including non-fumigatus Aspergillus species, Zygomycetes and Fusarium, have emerged, most invasive fungal infections (IFIs) are caused by Candida species and Aspergillus fumigatus.Citation1,Citation3

Predisposing factors for IFIs are well established. Specifically, modern chemotherapy for neoplastic diseases, with its immunosuppressive effects such as profound neutropenia, is a major risk factor.Citation1,Citation2,Citation4 The dysregulatory effects of antineoplastic agents on the inhibition of host leukocyte cell lineages and on other host immune responses that result in increased susceptibility to IFIs are well studied. Detailed descriptions of these immune modulatory effects are beyond the scope of this article but are comprehensively discussed elsewhere.Citation5Citation7 Interestingly, various anticancer agents, such as glucocorticoids, cytotoxic agents and calcineurin inhibitors, also have direct inhibitory effects on and/or have altered the biology of fungal cells.Citation8Citation11 Yet relatively little is known about these effects on the pathogenesis of IFIs and consequent clinical significance.

The advent of novel antineoplastic agents, such as the taxanes and inhibitors of histone deacetylase (HDAC), tyrosine kinase (TK) and heat shock protein 90 (Hsp90) complexes, has shifted cancer treatment paradigms from time-limited cytotoxic chemotherapy cycles to continuous low-dose treatment with protein-targeted agents.Citation12Citation14 Many targets of new anticancer agents have homologs in fungi and are components of critical cell-signaling pathways.Citation10,Citation11 Thus, studying new molecular targets and their inhibitors in the context of drug-pathogen interactions will provide insight into the pathogenesis of IFIs and may assist with antifungal drug development. This is pertinent given the current limited antifungal drug armamentarium.Citation1

In this article, we review current knowledge of the direct effects of conventional and investigational antineoplastic and/or immunosuppressive agents ( and ) on fungi. For each class of agent, we focus on the effects on fungal growth, morphological development and virulence, of the two most important pathogen genera, Candida and Aspergillus. Data on these fungi from direct in vitro and in vivo animal model-based studies of anticancer drugs, as well as from experiments using genetically engineered strains, are reviewed. We caution that these data are largely derived from “artificial” experimental systems, raising uncertainty about their application in the clinical setting. Nonetheless, understanding the fundamental in vitro and in vivo findings is an essential first step in informing the design of future studies that explore the clinical relevance of these observations where appropriate; such studies are currently lacking.

Conventional Cytotoxic Agents

Cytotoxic agents frequently used in cancer therapy include alkylating agents, antimetabolites, vinca alkaloids, anthracyclines (e.g., daunorubicin) and the antibiotic bleomycin () (also see section on DNA topoisomerase inhibitors). Alkylating agents exert their cytotoxic effects by adding alkyl groups to cancer-cell DNA; e.g., 5′-azacytidine (5-AC), a cytidine analog that specifically causes DNA hypomethylation. Antimetabolites such as cytosine arabinoside (ara-C) are structural analogs of naturally occurring competitive inhibitors of enzymes and molecules that inhibit progression to the S phase in DNA replication and, in some instances, inhibit DNA and RNA polymerases.Citation15 Vinca alkaloids inhibit microtubule aggregation.Citation16

Effects on Candida.

Antifungal activity. Cytotoxic agents have variable in vitro activity against Candida spp. (). In one report, the minimum inhibitory concentrations (MICs) of methotrexate, cyclophosphamide, vincristine, bleomycin and mitomycin C for three yeast strains (Candida albicans, Candida tropicalis and Candida kefyr) were high (500–1,500 µg/ml),Citation17 whereas those of asparaginase and 5-fluorouracil (5-FU) were less than 100 µg/ml. Both asparaginase and 5-FU were the most active against C. tropicalis (MIC, <1 µg/ml) suggesting Candida species-specific antifungal activity.Citation17,Citation18 In contrast, Graybill and colleagues noted that bleomycin was toxic in several Candida spp. in vitro, with minimum lethal concentrations ranging from 12.5 to 50.0 µg/ml (MICs, 0.39–12.5 µg/ml), but had no in vivo antifungal activity in mice with candidiasis.Citation19 It is unlikely that bleomycin results in clinically meaningful activity in vivo against Candida, as bleomycin is given once or twice weekly and in various doses with differing resultant plasma levels.Citation20 Indeed, the literature contains few data correlating the in vitro activity of cytotoxic agents with their in vivo efficacy. The anti-Candida activity of 5-FU is anticipated because 5-flucytosine, the less toxic precursor of 5-FU, is a well-established antifungal agent. Flucytosine is converted by cytosine deaminase to 5-FU in fungi, leading to blockage of protein synthesis. Unlike griseofulvin, which also inhibits fungal tubulin formation, the vinca alkaloids vincristine and vinblastine, even at clinically relevant concentrations, have little direct impact on fungal growth.Citation18

Studies of combinations of antifungal and antineoplastic agents have demonstrated inhibitory drug combinations whose MICs were lower than those for the respective agents alone.Citation17 In general, the combination of a polyene and an antineoplastic agent was the most potent inhibitor of growth of multiple Candida spp. Specifically, 5-FU had the most synergistic interaction with amphotericin B (AMB), but investigators observed similar interactions for AMB with methotrexate, mitomycin C and doxorubicin. However, cyclophosphamide and bleomycin antagonized AMB.Citation21 Again, the clinical relevance of these observations is not known. Large-scale testing of Candida strains across species using clinically relevant doses of various antineoplastic agents in clinically meaningful host models of infection is required to answer this question.

Morphology and virulence. Various effects of antineoplastic agents on Candida morphogenesis and virulence have been reported in vitro ().Citation22,Citation23 For example, both yeast-phase growth of and germ-tube formation by C. albicans were inhibited by treatment with actinomycin D, bleomycin, 5-FU and hydroxyurea at varying concentrations (100–1,000 µg/ml).Citation23 These effects were enhanced when the drugs were combined with AMB; in particular, actinomycin similar to the peak plasma concentrations of these agents that result from typical dosing; also, treatment with doxorubicin, vincristine and vinblastine inhibited Candida filamentation. These effects were potentiated when these drugs were combined with AMB; actinomycin D-AMB combinations resulted in 90% loss in cell viability.Citation23 In another study, at drug concentrations typically used during therapy, C. albicans isolates exposed to methotrexate but not cyclophosphamide, cytarabine or dacarbazine produced significantly higher amounts of catalase (a marker of oxitatative stress) than did unexposed isolates.Citation24 As catalase activity in Candida spp. is suggested to reduce the effects of AMB by protecting the species against oxidative damage,Citation25 methotrexate may contribute to AMB resistance.

In vitro exposure of Candida glabrata to high-dose methotrexate (500–1,500 µg/ml) also has inhibited the growth of and enhanced pseudomycelial formation by this species while restricting the uptake of RNA and DNA precursors into fungal into fungal cells.Citation26 Morphological alterations of Candida spp. following treatment with antineoplastic agents may explain the frequent isolation of morphologically-impaired isolates from patients with cancer.

Effects on Aspergillus.

The effects of ara-C and 5-AC on the nonpathogenic species Aspergillus nidulans are shown in .Citation27,Citation28 The “fluffy” phenotypes of A. nidulans and Aspergillus niger are characterized by severely impaired asexual sporulation, abundant vegetative hyphae and uncontrolled growth.Citation29 Similarly, exposure of an A. fumigatus strain to high concentrations (250–500 mM) of 5-AC induced high-frequency conversion of the strain to a developmental mutant with impaired light-dependent conidiation accompanied by increased elastase and proteolytic activity, greater angioinvasion in vitro than that in the wild-type strain, and overexpression of genes encoding for cell wall-associated proteins. Mutants of this type remained pathogenic in fruit fly and murine model systems.Citation30 However, the concentrations of 5-AC required to induce the fluffy phenotype exceeded those achievable in humans during treatment.Citation31

Metal Complexes

The platinum compounds cisplatin, carboplatin and transplatin; arsenic trioxide (ATO); and zinc-containing agents have emerged as clinically useful metal complexes in the treatment of cancer (). Data on their effects on the growth and/or development of fungi, however, are relatively lacking.

Cisplatin.

Effects on Candida. Researchers have studied the effects of cisplatin on a number of yeasts, including Saccharomyces cerevisiae and C. albicans with variable results.Citation11 One study showed that cisplatin inhibited C. albicans growth only at concentrations much higher (100–800 µg/ml) than those achievable in vivo,Citation32 whilst another using the strain C. albicans ATCC 10261 noted persistent (up to 40 h) inhibition at concentrations as low as 20 µg/ml.Citation33 Serum concentration exposures of cisplatin vary according to the underlying malignancy. In testicular cancer cases, plasma levels of cisplatin equivalent to a Cmax of 1.95 µg/ml are typical targets.Citation34 In addition to causing aberrant cross-linking of host DNA strands, cisplatin inhibits rRNA synthesis as well as the self-splicing reaction of the group-I intron ribozyme of pre-rRNA in mammalian cells.Citation35 Although C. albicans possesses this group I self-splicing intron, whether cisplatin exerts its inhibitory effect via this mechanism is not known. Pretreatment of C. albicans with AMB (and miconazole) has increased its sensitivity to cisplatin in vitro, suggesting a degree of synergistic antifungal activity of these agents (),Citation17,Citation36 but these observations require confirmation.

In studying changes in the morphology and growth of C. albicans using fluorescence microscopy, Chandrasekar et al.Citation37 showed that cisplatin exposure (6 µg/ml) markedly inhibited hyphal and bud development. Also, C. albicans cells exhibited increased uptake of ethidium bromide and were unusually large before dying. These changes were concomitant with suppression of growth of cisplatin-exposed C. albicans compared with untreated cells. In addition, cisplatin may potentiate the virulence of Candida cells. In one study, cells from two Candida strains cultured in the presence of 10–100 µg/ml cisplatin proliferated more rapidly than did untreated cells,Citation9 demonstrated increased adhesion to HeLa cells via lectin binding, had increased enolase and acid proteinase activity, and were more resistant to neutrophil killing than were unexposed to cisplatinum C. albicans control cells. The researchers also observed similar results for 5-FU (250 µg/ml). In contrast with previous findings, however,Citation17,Citation36 cisplatin decreased the sensitivity of Candida cells to AMB when the half-maximal inhibitory concentration of this agent was increased 1.5- to 2.0-fold.Citation9 The clinical implications of these effects have not yet been investigated.

Effects on Aspergillus. At the time of this writing, whereas investigators have shown that cisplatin promotes mitotic recombination in A. nidulans, no reported data have indicated that this occurs in A. fumigatus ().

Arsenic (ATO).

Despite the fact that arsenic is considered a poison, ATO is a valuable treatment option for cancer, especially acute promyelocytic leukemia. Although its predominant antineoplastic activity occurs via induction of apoptosis, ATO also induces intracellular activity of reactive oxygen species, activates enzyme kinases, and is involved in inhibition of P-glycoprotein and potentiation of tubulin polymerization.Citation38

Effects on fungi. Genome-wide analyses of transcription patterns in S. cerevisiae in response to treatment with arsenic have revealed a complex response that influences signaling pathways including protein kinases such as the mitogen-actived protein kinase and target of rapamycin (TOR) complex 1 (TORC1) systems.Citation39,Citation40 Arsenic toxicity results from inhibition of TORC1 kinase and modulation of the activity of downstream factors, leading to activation of the transcription factors that are targets of TORC1 and protein kinase A.Citation40 Whether arsenic has similar effects in Candida and Aspergillus spp. is unknown. In other studies, A. niger isolates recovered from arsenic-rich environments demonstrated reduced radial growth (∼50%), formed tiny black compact colonies with dense sporulation and exhibited greater catalase activity than did laboratory control strains.Citation41,Citation42 The relevance of these in vitro findings is unknown.

Taxanes

The diterpene taxanes are also integral parts of modern cancer treatment regimens.Citation12,Citation43 They reorganize and stabilize cell microtubules, interfering with their normal breakdown during cell division.43 Licensed agents include paclitaxel, docetaxel and carbazitaxel and less toxic nanoparticle albumin-bound formulations (e.g., nab-paclitaxel) and paclitaxel derivatives (e.g., bound to a polyglutamate polymer, PG-paclitaxel).Citation44 Taxanes are produced by several fungi,Citation45 yet in vitro (as well as in vivo) data regarding their direct effects on the biology of pathogenic fungi are scarce. Possible effects may be assessed in the context of studies of S. cereviase. In wild-type S. cerevisiae, paclitaxel does not bind to fungal tubulin. However, S. cerevisiae strains with mutated β-tubulin genes leading to diminished transport activity of tubulin exhibit evidence of mitotic blockage with cell apoptosis after exposure to paclitaxel.Citation46 The microfilament physiology in of C. albicans is known to play a critical role in germ-tube formation during pH-regulated morphological yeast to hyphal transition.Citation47 However, whether taxanes administered in pharmacological doses (e.g., typical paclitaxel doses of 135–175 mg/m2 result in plasma Cmax values of 0.195–0.355 µg/ml) affect microtubule formation in vitro or in vivo is unknown. Although the cytoskeletal inhibitors cytochalasin A and nocodazole disrupted microfilament formation in endothelial cells and subsequently impaired phagocytosis of C. albicans in one study, the pattern of actin polymerization-driven phagocytosis of C. albicans by endothelial cell was unaffected by treatment with 1 µM paclitaxel.Citation48

Corticosteroids

The vulnerability to IFIs of patients with cancer receiving longterm glucocorticoid-based therapy (e.g., dexamethasone, hydrocortisone, methylprednisolone) at supraphysiological doses is attributed to dysregulation of macrophage and polymorphonuclear function by excessive plasma cortisol.Citation5 Treatment with steroids reduces the ability of innate effectors to kill microorganisms via multiple mechanisms, including production of reactive oxygen species. Data on the direct effects of corticosteroids on fungi are summarized in .

Effects on Candida.

Early observations that treatment with glucocorticoids increased the C. albicans burden in the gastrointestinal tract and frequency of fungal translocation from the gastrointestinal tract to the bloodstream in mice and rats,Citation49,Citation50 raised the possibility that steroids can augment the growth rate of Candida spp.Citation5,Citation51 Their direct effects on Candida growth per se however, remain undetermined.

Nonetheless, investigators have shown that glucocorticoids enhance adhesion of Candida spp. to vaginal epithelial cells.Citation52 Also, treatment with dexamethasone (0.01% w/v) increased adhesion (by 17–44%) of these species to buccal epithelial cells, although that with cortisone acetate and hydrocortisone (both at 0.01% w/v) decreased the adhesion by 16–32%.Citation53 In another study, exposure to glucocorticoids stimulated secretion of Candida phospholipase and aspartyl proteinase,Citation54 whereas Gyetvai et al. found that high-dose treatment (4 mM) with methylprednisolone stimulated C. albicans growth, the germination rate in serum and phospholipase release from C. albicans. Surprisingly, these effects were not accompanied by increased pathogenicity in intraperitoneal and intravenous mouse models of infection. Methylprednisolone-based treatment also increased the sensitivity of C. albicans cells to menadione via inhibition by inhibiting antioxidant enzymes and enhanced the deleterious effects of AMB.Citation55 Adhesion to human epithelial cells and secretion of extracellular proteinases and phospholipases are virulence traits of C. albicans.Citation56

The mechanisms by which corticosteroids affect Candida biology and growth are not well understood. Researchers identified a corticosteroid-binding protein (CBP) on the surface of C. albicans cells.Citation57 Investigation is warranted to determine whether steroid-CBP binding enhances Candida virulence. Of interest, treatment with ketoconazole, at concentrations readily achievable in therapeutic settings, has displaced corticosterone from the CBP site in C. albicans.Citation58 Genome-wide analyses of C. albicans cells treated with the human steroid progesterone have identified enhanced expression of multidrug resistance genes of the ATP-binding cassette transporter [Candida drug resistance 1 (CDR1) and CDR2] superfamily, suggesting the presence of a relationship between steroid-induced stress and azole resistance in C. albicans.Citation59 The expression of several genes associated with induction of hyphal growth and establishment of pathogenesis in C. albicans was upregulated with specific progesterone-responsive sites in the promoter regions of these genes.Citation59 In a detailed study of the C. albicans CDR1 promoter, researchers identified two cisregulatory regions responsive to progesterone and β-estradiol, respectively: steroid responsive element (SRE)1 and SRE2.Citation60 Whether glucocorticoids in clinical use have similar roles in regulation of the expression of CDR1 and other genes in C. albicans remains to be elucidated.

Effects on Aspergillus.

Treatment with glucocorticoids may also alter the biological fitness of Aspergillus spp. (). Ng et al. first showed that in vitro exposure to pharmacological doses of hydrocortisone increased the growth (by 30–40%) of A. fumigatus and Aspergillus flavus.Citation8 This direct growth effect may potentiate the immunosuppressive action of hydrocortisone, facilitating dissemination of Aspergillus in vivo.Citation5 Interestingly, treatment with progesterone and estradiol had no effects on A. fumigatus growth.Citation8 Since many patients with cancer simultaneously receive glucocorticoids and antifungal drugs, the combined effects of these agents on Aspergillus growth would be a relevant topic of study. In another study, treatment with a therapeutic concentration (1 µM) of hydrocortisone induced a significant increase in susceptibility to itraconazole but not to AMB in A. fumigatus ().Citation61 The mechanisms of these interactions, and whether the interactions are clinically meaningful, are not known.

Inhibitors of Calcineurin and TOR

Agents that inhibit the Ca2+/calmodulin-activated protein phosphatases calcineurin and TOR are the cornerstones of therapy in patients who undergo organ or bone marrow transplantation as well as chemotherapy for cancer. Despite having intrinsic antifungal activity, the development of currently-available agents as anti-infective drugs has been limited by their potent immunosuppressive effects. Indeed, the relative high rates of IFIs in patients receiving these agents emphasise the complex interplay of host and pathogen-specific factors that influence the composite host response to any one drug.

The relevance of calcineurin and TOR to fungal biology relates to the central roles of (1) TOR kinase and (2) calcineurin and its modulator Hsp90 in regulating cell growth and stress responses. Calcineurin is essential for fungal morphogenesis and virulence and positively regulates cell-wall synthesis through in FKS2 gene expression via the calcineurin-regulated zinc fingercontaining protein (Crz1p) or its homologs.Citation10,Citation62,Citation63

Inhibitors of these conserved targets—cyclosporin A (CsA), tacrolimus (FK-506; calcineurin), rapamycin (sirolimus; TOR), Hsp90 inhibitors, and their less immunosuppressive analogs have become foci of study for their effects on fungal development (). Key findings have revealed interplay among TOR, calcineurin and Hsp90 inhibitors in regulating polarized growth and stress responses of fungi, which have opened up the possibility of exploiting these pathways to design novel synergistic combination therapies between conventional antifungals and inhibitors of these targets (). Expert reviews have detailed these signaling cascades.Citation10,Citation62,Citation64

Lessons from studies in S. cerevisiae.

Researchers first identified the TOR kinases as in S. cerevisiae and showed that rapamycin forms a complex with the tacrolimus-binding protein FKBP12 prolyl isomerase, which then binds to TOR kinase, blocking its function. Although S. cerevisiae contains two TOR complexes (TORC1 and TORC2), other fungi have single TOR homologs.Citation10 Rapamycin inhibits ribosome synthesis, mating and filamentation and induces autophagy in S. cerevisiae.Citation65 In vitro, rapamycin MICs range from 0.1 to 5.0 µg/ml, whereas CsA and tacrolimus MICs are 10- to 1,000-fold higher.Citation66

Effects on Candida.

The advent of less immunosuppressive analogs of the calcineurin pathway (CsA, tacrolimus) and the lipid-formulated rapamycin, and the finding that the aforementioned compounds are synergistic in vitro with AMB and 5-flucytosine against Candida spp. renewed interest in their potential antifungal activity in the clinic.Citation67 Prior studies established that rapamycin and its analogs (particularly analogs 23 and 2) were fungicidal against C. albicans because of FKB12-dependent inhibition of TOR; the rapamycin MICs in these studies ranged from less than 0.02 to 0.20 µg/ml with an MFC of 0.39 µg/ml.Citation66,Citation68 At sublethal concentrations, rapamycin blocked C. albicans filamentation via expression of genes associated with nitrogen use, such as mep2 and gln3.Citation10,Citation65,Citation69 Mutants defective in these genes were avirulent in mice with disseminated candidiasis.Citation69 In another study, oral administration of 10 and 12.5 mg/kg rapamycin protected 50% and 90%, respectively, of mice against death from C. albicans infection.Citation70

Despite having high MICs (>10–100 µg/ml) in Candida spp.,Citation66 both tacrolimus and CsA (which complex with FKBP12 and cyclophilin, respectively) may influence the efficacy of antifungal drugs and reduce C. albicans virulence ().Citation62,Citation71,Citation72 These in vitro results are supported by gene deletion studies showing that calcineurin-defective C. albicans mutants were more sensitive to stress conditions than isogenic wild-type controls, were markedly hypersensitive to azoles, and had attenuated virulence in mice with systemic candidiasis.Citation73Citation75 Although that hypovirulence is attributed to sensitivity of C. albicans to killing by serum mediated by Ca2+, no reported data have indicated that pharmacological inhibition of calcineurin also protects against calcium stress. Interestingly, loss of function crz1 C. albicans mutants, defective in crz1 which encoded a downstream transcription effector of the calcineurin, Crz1, were not sensitive to serum and exhibited no decrease in virulence.Citation76

Synergistic interaction among CsA, tacrolimus and fluconazole in vitro () is attributed to conversion of the static effect of fluconazole to fungicidal activity in Candida species. This interaction extends to the other azoles as well as terbinafine and was observed in both susceptible C. albicans and azole-resistant species, including C. glabrata.Citation71Citation73 Checkerboard microdilution and time-kill experiments found that at a concentration of 0.625 µg/ml (i.e., at a concentration achievable in vivo), CsA was synergistic with fluconazole, reducing viable Candida counts by 2 logs.Citation77 Importantly, testing of Candida isolates obtained from liver transplant recipients receiving tacrolimus showed that treatment with this agent did not select for resistance toward calcineurin inhibitors.Citation78

In general, these data are supported by the results of in vivo animal studies. Treatment with a CsA-fluconazole combination (at therapeutic drug levels) was more effective than that with either drug alone against experimental candidal endocarditis and in mice with Candida keratitis.Citation77,Citation79 In a rat study, C. albicans embedded in biofilm and was resistant to fluconazole, CsA and tacrolimus alone but highly susceptible to CsA-fluconazole and tacrolimus-fluconazole combinations.Citation80 However, the influence of calcineurin on C. albicans virulence appears to be host- and niche-specific. Investigators observed that calcineurin-defective C. albicans strains were hypovirulent in mice with systemic or ocular infections but fully virulent in mice with lung, vaginal or oropharyngeal infections.Citation10,Citation74,Citation75 Such C. albicans mutants may be unable to survive serum-induced calcium stress, accounting for their reduced virulence in systemic infections. Recently, a C. glabrata calcineurin mutant, defective in the calcineurin regulatory B subunit, exhibited increased susceptibility to both azoles and micafungin and was hypovirulent in mice. Unlike with C. albicans, loss of the C. glabrata homolog Crz1 did not result in increased azole susceptibility.Citation81 The in vitro and in vivo (in animals) synergy of CSA/tacrolimus and azoles is intriguing; both classes of drug may be immuno suppressive again emphasizing the need to study host as well as and pathogen-specific factors in predicting likely response of a pathogen to any therapeutic agent.Citation82,Citation83

Reported data on the effects of CsA and tacrolimus on Candida morphogenesis are few. In one study, CsA administered at concentrations of 50–1,000 µg/ml inhibited germ-tube induction (50% inhibition at 100–500 µg/ml) and mycelial transformation of blastospores in C. albicans ().Citation84

Effects on Aspergillus.

Extensive studies of A. fumigatus indicated that calcineurin has a critical role in regulating polarized hyphal growth of and tissue invasion by this species.Citation62 In A. nidulans for instance, cnaA, the gene that encodes calcineurin, is essential for conidial germination. CnaA mutants in A. fumigatus animal models exhibited defective conidial and hyphal extension and significant attenuation of pathogenicity (mortality rate, 10% vs. 90%); hyphae were almost completely absent from lung tissue.Citation62,Citation84 cnaA A. fumigatus mutants defective in the calcineurin A unit also demonstrated aberrant hyphal growth and reduced pathogenicity.Citation85,Citation86

Direct evidence of the effects of calcineurin and TOR inhibitors on the phenotype of Aspergillus spp. () include reports that rapamycin inhibited the growth of A. fumigatus, A. flavus and A. niger in vitro (MICs, ∼50 µg/ml) but that this inhibition was not sustained.Citation87,Citation88 In other studies, CsA and tacrolimus had intrinsic activity against A. fumigatus (CsA: MICs, 3–10 µg/ml; geometric mean MIC, 6.25 µg/ml; tacrolimus: MICs, 0.2–0.4 µg/ml; geometric mean MIC, 1.56 µg/ml).Citation66,Citation89 Calcineurin and TOR inhibitors also have converted azoles (e.g., itraconazole) and the echinocandin caspofungin into fungicidal compounds against A. fumigatus, A. flavus and Aspergillus terreus.Citation87Citation89 Inhibition of the calcineurin and TOR pathways may decrease the threshold of deleterious effects of echinocandins. Alternatively, combinations of caspofungin and calcineurin inhibitors may inhibit expression of the A. fumigatus FKS1 homolog.Citation89 As for Candida spp., long-term CsA and tacrolimus exposure in vivo did not make these species resistant to these agents.Citation88

Animal data have indirectly confirmed the CsA- and tacrolimus-induced anti-Aspergillus effects described above in vivo. For example, a study showed a 100% mortality rate in granulocytopenic rabbits given ara-C but a 100% survival rate in those given CsA and methylprednisolone.Citation90 In another, A. fumigatus model, survival rates were significantly higher and survival durations were significantly longer in mice receiving low-dose (1 mg/kg/day) intravenous tacrolimus but not CsA. Also, researchers observed numerous Aspergillus hyphae in the brains and kidneys of CsA-treated mice but no hyphae in the brains of tacrolimus-treated animals; it is unclear if these observations are directly related to an in vitro drug effect and/or from effects on the host immune systemCitation91 and again, their relevance (or not) in the clinical setting remains to be established.

Tacrolimus and CsA exhibit differences in their effects on the morphology of Aspergillus spp. (). For example, authors reported gross stunting of the growth with blunting of hyphal tips after tacrolimus but not CsA exposure.Citation88,Citation92 However, administered alone, both agents delayed filamentation of A. fumigatus, which was enhanced when either agent was given with caspofungin. Quantification of cell wall 1,3-β-glucan synthase activity revealed decreased levels of activity after calcineurin inhibition, supporting the hypothesis that calcineurin inhibition operates via cell-wall biosynthesis.Citation93 Identification of fungus-specific downstream effectors of calcineurin in fungi may lessen any broad cross-reactivity of current calcineurin inhibitors with human targets. A study showed that the CrzA homolog in A. fumigatus (not found in mammals) was essential for the growth, germination, sexual development and pathogenicity of A. fumigatus.Citation94 Inhibition of CrzA is worthy of consideration as one of the next generation of calcineurin inhibitors.

Effects on other fungi.

Although beyond the scope of this review, rapamycin and the calcineurin inhibitors have inhibited the growth of other fungi, most notably Cryptococcus neoformans.Citation62 The calcineurin pathway is integral to cryptococcal pathogenesis and parallels regarding other pathogenic fungi may be drawn from erported data on this pathogen.Citation62,Citation95

Hsp90 Inhibitors

The molecular chaperone Hsp90 is part of a multichaperone complex essential for the conformation and function of a diverse set of client proteins, including key oncogenic proteins, calcineurin, TKs and serine/threonine kinases, many of which regulate cell signaling and stress response.Citation13,Citation96,Citation97 ATPase activity combined with ADP/ATP exchange in this complex is essential for the control of client-protein binding and fate and involves the association and/or dissociation of co-chaperones (e.g., Hsp40, Hsp90/70 organizing protein) Pharmacological inhibitors of Hsp90 typically dock in the N-terminal nucleotide binding site with high avidity, interrupting ATPase activity and resulting in downstream degradation of client proteins.Citation13

The first-in-class Hsp90 inhibitor tested in clinical trials for cancer treatment was the ansamycin antibiotic geldanamycin analog 17-allylamino-17-demethoxygedanamycin (17-AAG; geldanamycin itself is too toxic for clinical use) administered either as a single agent or in combination with cisplatin and paclitaxel. Other Hsp90 inhibitors include another geldanamycin analog, 17-desmethoxy-17-N,N-diemthylaminoethylaminogeldanamycin (17-DMAG); the oxime derivatives of radicicol; pyrazoles; purines; novobiocin analogs; and inhibitors of Hsp90 isoforms, including a recombinant antibody against fungal Hsp90 (described below). Cisplatin, which binds to the Hsp90 C-terminal domain; paclitaxel; and the HDAC inhibitors (HDACis), which induce acetylation of Hsp90 (described below), also decreases Hsp90 function.Citation13,Citation96,Citation97 Hsp90 inhibitors are already known to inhibit viral replication.Citation98 Because Hsp90 is highly conserved in eukaryotes, their pharmacological inhibition may affect fungal development.

Effects on Candida.

Researchers first obtained data linking Hsp90 with the emergence of antifungal drug resistance () in S. cerevisiae.Citation99 Similarly, investigators have shown that the Hsp90-calcineurin signaling network plays a key role in the rapid development and maintenance of C. albicans resistance to azoles as well as the echinocandins (). At concentrations well tolerated in humans, treatment with radicicol prevented the emergence of resistance to azoles and abrogated the resistance of laboratory mutants of C. albicans as well as C. albicans strains in which resistance to azoles developed in a human host.Citation99,Citation100

More recently, 17-AAG and 17-DMAG administered at concentrations with minimal direct activity against C. albicans were synergistic in vitro with fluconazole.Citation101 The efficacy of fluconazole with these two Hsp90 inhibitors in an invertebrate model (larvae of the wax moth Galleria mellonella) was greater than that when it was administered alone as the combination therapy rescued larvae from lethal infection with C. albicans. That study also showed fungicidal activity of azoles when administered along with geldanamycin in viability assays and in mice with candidiasis.Citation101 Singh and colleagues further showed that pharmacological (geldanamycin) and genetic impairment of Hsp90 function reduced the micafungin tolerance of laboratory C. albicans strains and micafungin resistance of clinical C. albicans isolates, resulting in a fungicidal combination of micafungin and geldanamycin.Citation102 Investigators have expressed concerns with the potential for reduced activity of echinocandins at high concentrations, known as the “paradoxical” effect. In one study, treatment with radicicol reduced the paradoxical effect of micafungin in vitro, reduced tolerance to voriconazole and abrogated micafungin-voriconazole antagonism in C. albicans.Citation103 Intriguingly, Hsp90 inhibition-induced filamentation of C. albicans via regulation of cyclic adenosine monophosphate-dependent protein kinase A signaling, and may be relevant to the virulence of C. albicans.Citation104

Further, studies have shown that C. albicans Hsp90 is an immunodominant antigen in mice and humans. For example, researchers tested a recombinant C. albicans-specific anti-Hsp90 antibody (efungumab) to determine its antifungal activity.Citation105,Citation106 Although Candida isolates display high efungumab MICs of 128–256 µg/ml, efungumab was synergistic with both AMB and caspofungin against Candida against these isolates. Furthermore, a clinical trial of invasive candidiasis demonstrated a therapeutic benefit of efungumab when administered with AMB.Citation107 Efungumab likely affects the host immune responses to the pathogen because the antibody is unable to cross the fungal cell wall and enter the cytosol, where Hsp90 inhibitors exert their activity.Citation108 Overall, given the promising in vitro results obtained with the HSP90 inhibitors, further studies systematically exploring the clinical efficacy, particularly of enfungumab formulations, are warranted to determine their position in the anti-Candida drug armamentarium.

Effects on Aspergillus.

Unlike C. albicans, little is known about the direct effects of Hsp90 inhibitors on the development of drug resistance in Aspergillus spp., although a recent study confirmed that Hsp90 is required for basal resistance of these species to the echinocandins.Citation101 Treatment with geldanamycin has enhanced the activity of caspofungin against single strains of A. fumigatus as well as against A. terreus.Citation99,Citation101 Inhibition of Hsp90 enhanced the activity of voriconazole against A. fumigatus, although this effect depended on environmental conditions.Citation101 Finally, combination treatment with geldanamycin and caspofungin rescued G. mellonella larvae from lethal infection with A. fumigatus.Citation101

HDACis

HDACs are chromatin-modeling enzymes that deacetylate lysine residues on core histones and other cell proteins. Central to gene regulation, HDACs control a range of cellular functions, including proliferation and death. HDACis thus modulate the acetylation status of chromatin and a range of cellular proteins, including Hsp90, leading to transcriptional cell-signaling events that induce growth arrest, apoptosis and autophagy (described below). HDACis exhibit promise in the treatment of cancer and certain parasitic and viral infections.Citation14,Citation109

The HDACis include trichostatin A (TSA), romidepsin/depsipeptide, suberoylanilide hydroxamic acid (SAHA; vorinostat), panobinostat and MG-CD0103 (mocetinostat). However, only SAHA and romidepsin are licensed for clinical use (for cutaneous T cell lymphoma).Citation14,Citation110,Citation111 As HDACis potentiate the cytotoxicity of DNA methylation inhibitors (e.g., cisplatin, the anthracyclines, dexamethasone), they may be used together with these inhibitors.Citation109Citation111 Mammalian and fungal HDACs are substantially homologous and investigators have identified several classes and subclasses of HDACs in fungi (e.g., the class I enzymes RpdA and HosA, the class II enzymes HdaA and HosB).Citation112 Amongst proteins within the HDAC complex, H3K56ac has a key role in the DNA damage response; this protein may be fungal-specific.Citation113

Effects on Candida.

C. albicans have two distinct cell types—white and opaque—with phenotypic differences in cell shape, mating properties and virulence. White cells are typically found in the bloodstream, whereas opaque cells are mainly located on mucosal surfaces and skin. HDACs regulate these phenotypes.Citation14 Treatment with TSA can increase the occurrence of white-to-opaque cell transition by inhibiting the expression of HDAC genes HDA1 and RPD3 ().Citation114 TSA may also impact azole susceptibility in Candida spp. In one study, treatment with TSA at concentrations ranging from 2 to 3 µg/ml markedly reduced the azole trailing effect and enhanced azole susceptibility by reducing the upregulation of expression of the CDR and ERG genes in a small number of strains of C. albicans, C. tropicalis and Candida parapsilosis but not C. glabrata.Citation115 Conversely, treatment with TSA at 1–32 µg/ml had a minimal effect on in vitro growth of, germ-tube formation by (in 20% serum) and heat sensitivity of these strains,Citation115 although another study found that both TSA and SAHA inhibited serum-induced germ-tube formation in C. albicans.Citation116 In combination with fluconazole, treatment with several investigational uracil-based hydroxamates inhibited induction of resistance to fluconazole in Candida spp.Citation117 TSA and SAHA also caused a 90% reduction in C. albicans adherence to human cultured pneumocytes.Citation118 Increasing experience in using HDACis to treat cancer will aid testing of the efficacy of combinations of HDACis and azoles against Candida spp. at pharmacologically relevant concentrations and in in vivo models of infection.

Novel HDACis with activity against Candida HDACs, particularly those selective for HDAC6 and Hda1p, are worthy of investigation as next-generation HDACis because of their potential antifungal activity.Citation118 The protein Hda1p appears to play a key role in development of drug resistance in Candida spp. as such resistance cannot develop in HDA1-deficient strains.Citation14 In addition, data have shown that HDAC6 regulates Hsp90 acetylationCitation119 and that hyperacetylation of the Hsp90 complex leads to inhibition of it. Finally, reduced levels of H3K56ac activity (regulated by the C. albicans RTT109 gene) have sensitized C. albicans to hydroxyurea, the azoles, and echinocandins and are associated with attenuated C. albicans virulence in mice.Citation113,Citation120 Inhibition of Hst3p in C. albicans results in loss of cell viability with abnormal filamentous growth.Citation113 Researchers have tested the activity of at least one new HDACi, MG-CD290, against a range of Candida spp. and molds (described below; ).Citation121

Effects on Aspergillus.

The effects of HDACis on A. nidulans are summarized in . Inhibition of HDACs targets primarily the class II enzyme HdaA, which accounts for the majority of HDAC activity in Aspergillus.Citation122 Depletion of the RPD3-type HDAC RpdA in A. nidulans leads to a pronounced reduction in growth and sporulation of this species.Citation123

Pfaller et al.Citation121 examined the antifungal activity of MGCD290, a novel Hos2 fungus-specific HDACi, in combination with fluconazole, voriconazole and posaconazole against a range of yeasts (including Candida; n = 30) and molds (including Aspergillus; n = 10). MGCD290 MICs ranged from 0.5 to 16.0 µg/ml for Candida spp., including azole-resistant strains, whereas the MICs ranged from 8 to >32 µg/ml for Aspergillus spp. Notably, MGCD290 (8–32 µg/ml) was synergistic with fluconazole against 87% of Candida isolates and unexpectedly enhanced fluconazole activity against six Aspergillus isolates. Importantly, in Aspergillus spp., MGCD290 was synergistic with voriconazole (six isolates) and posaconazole (four isolates). Current studies are examining optimal dosing of MGCD290 with phase I first-in-human clinical studies demonstrating safety and favorable pharmacokinetic properties of this agent when administered alone or with fluconazole (www.methylgene.com).

DNA Topoisomerase Inhibitors

That DNA topoisomerase inhibitors directly affect the growth and biology of fungi is not a new concept.Citation124 These compounds include the fluoroquinolones, anthracyclines and bleomycin.

Fluoroquinolones.

Fluoroquinolones inhibit DNA gyrase (topoisomerase II) and topoisomerase IV, resulting in cell death. Topoisomerases are universally present in eukaryotes, with high levels of topoisomerase I and II in fungi.Citation124 Data have indicated the potential of fluoroquinolones, especially ciprofloxacin, ofloxacin and moxifloxacin, to enhance the efficacy of antifungal drugs despite lacking intrinsic antifungal activity.Citation124,Citation125

Effects on Candida. Assessment of in vitro pharmacodynamic interactions between antifungal and nonantifungal agents is complicated by the absence of a common antifungal end point using conventional fractional inhibitory concentration index criteria. However, recent in vitro isobolographic analyses showed that significant pharmacodynamic interactions between ciprofloxacin and AMB and between voriconazole and caspofungin do occur in C. albicans.Citation126,Citation127 Specifically, AMB and ciprofloxacin were synergistic at ciprofloxacin concentrations less than 10.64 µg/ml and AMB concentrations less than 0.33 µg/ml but may be antagonistic at higher ciprofloxacin concentrations.Citation126 Other fluoroquinolone-AMB and fluoroquinolone-azole interactions vary according to the fluoroquinolone ().Citation127 Variable interactions between fluoroquinolones and caspofungin also have occurred, with indifferent to synergistic results.Citation126 Fluoroquinolones may inhibit fungal DNA replication, but this effect is apparent only when combined with antifungal drugs. Also, increased cell-membrane permeability resulting from treatment with antifungal drugs may increase intracellular ciprofloxacin levels, leading to synergy of the antifungal drugs and this fluoroquinolone. Alternatively, ciprofloxacin may interact with cholesterol-containing liposomesCitation128 and participate in pore formation in fungal cell membranes, thus enhancing AMB activity.

The above in vitro observations are supported by limited animal data—synergistic interactions of ciprofloxacin plus fluconazole, ciprofloxacin plus AMB and fluconazole plus ofloxacin in vivo in mice with C. albicans infection are described.Citation129 However, liposomal AMB and moxifloxacin were not synergistic in a rabbit model of C. albicans endophthalmitis for this species.Citation130,Citation131 In another study, moxifloxacin protected cyclophosphamide-injected mice against C. albicans bronchopneumonia and reduced mortality rates.Citation132 Yet, long term quinolone use is known to be a risk factor for Candida infectionCitation1 emphasizing the complexity of drug-pathogen interaction in a specific host environment and the need to study fungal pathogenesis in appropriate clinical contexts.

Effects on Aspergillus. Likewise, researchers have observed synergistic interactions of AMB with ciprofloxacin and levofloxacin (but not moxifloxacin) in A. fumigatus.Citation126,Citation127 Combinations of ciprofloxacin with voriconazole and caspofungin were also synergistic in these studies (). For ciprofloxacin-AMB combinations, investigators detected synergy at low concentrations of both ciprofloxacin (0.78–1.85 µg/ml) and AMB (0.24–0.39 µg/ml) but noted antagonism at higher concentrations.Citation126 Animal models of invasive aspergillosis have supported the enhanced efficacy of antifungals when combined with fluoroquinolones.Citation129 Genus-specific differences between Candida and Aspergillus in the interactions of fluoroquinolones with individual antifungals may be related to different affinities of each quinolone for fungal topoisomerases.Citation127 Determing the clinical relevance (or lack thereof) of these experimental interactions is indicated.

Anthracyclines.

The anthracycline antibiotics induce faulty retyping of DNA strands by impairing topoisomerase II activity. Doxorubicin (Adriamycin) also affects mitochondrial function by selectively binding to cardiolipin in the mitochondrial cell membrane.Citation133 Other anthracyclines are daunorubicin, aclarubicin and idarubicin.

Effects on Candida and Aspergillus. In studies of simulated blood cultures, doxorubicin inhibited the growth of C. albicans, whereas at physiologically achievable concentrations, idarubicin inhibited the growth of C. glabrata but not C. albicans or Candida krusei.Citation134,Citation135 In another study, aclarubicin displayed fungistatic activity against C. albicans (MIC range, 0.8–7.3 µg/ml), but doxorubicin, idarubicin and daunorubicin had much higher MICs (>26 µg/ml); furthermore, idarubicin inhibited hyphal formation by C. albicans.Citation136 Exposure of C. albicans to doxorubicin resulted in mitochondrial dysfunction, decreased in ergosterol content in fungal cell membrane (ergosterol is the target binding site of AMB), all culminating in increased the tolerance of this species to AMB.Citation137 Most recently, Kofla et al. reported that doxorubicin induced CDR1/CDR2 expression in C. albicans at both the mRNA and protein level and caused a 4–8-fold increase in MICs to fluconazole.Citation138 Expression of the multidrug resistant 1 (MDR1) and ERG11, genes were not affected.Citation138 Data on the effects of anthracyclines on Aspergillus spp. are limited ().Citation139

Bleomycin.

The glycopeptide bleomycin induces oxidative-mediated DNA damage manifested by double- and single-strand DNA breaks and mediates RNA degradation at sites adjacent to guanosine residues.Citation140

Early studies of mice with candidiasis demonstrated activity of bleomycin against C. albicans ().Citation19 Recently, a study showed that bleomycin inhibited self-splicing of the 25S rRNA Group I intron of C. albicans at a half-maximal inhibitory concentration of 1.2 µM, resulting in accumulation of RNA precursors and inhibition of C. albicans growth by about 44%.Citation140 The intron-containing C. albicans strain had a bleomycin MIC of 1.56 µg/ml, whereas the intronless strain had a MIC of 6.25 µg/ml. This difference in MICs is thought to be related to selective interaction of bleomycin with folded RNA structures.

Bleomycin also can impact the phenotype of A. nidulans ().Citation141 Researchers have observed direct activity of bleomycin against A. fumigatus (MIC, 3.2 µg/ml) with arrest of conidial germination, prevention of hyphal development and completion of cell-wall septation.Citation142 A possible mechanism of cell-wall damage caused by bleomycin is increased cell-wall permeability via disruption of glycosidic cross-linking structures.Citation142

Other antibiotics.

The sulphonamides, rifampicin and metronidazole have also been studied alone or in combination with polyenes or azoles in vitro and in animal models for their antifungal effects. Sulphamethoxozle drugs were found to be active against most Aspergillus species and may help to prevent invasive aspergillosis in AIDS patients (reviewed in ref. Citation143). Ketoconazole-sulfamethoxazole combinations were synergistic against C. albicans.Citation144 Rifampicin, a macrocyclic antibiotic in combination with AMB has synergistic interactions against certain Candida and Aspergillus spp.Citation143 Likewise, additive to synergistic effects have occurred between AMB and metronidazole.Citation145 The clinical implications of these observations have yet to be determined.

TK Inhibitors

TK inhibitors (TKIs) target enzymes whose dysregulated expression and activity are associated with various cancers. As such, small-molecule TKIs that prevent autophosphorylation and propagation of downstream intracellular signals and monoclonal antibodies against TKs that block ligand binding have revolutionized the treatment of certain cancers.Citation146,Citation147 Small-molecule TKIs compete with ATP binding to the TK domain and abrogate the catalytic activity of the TKI receptors but may also be “non-receptor-targeted” (non-receptor TKIs, e.g., the src TK family).Citation147 These include imatinib mesylate (a nonreceptor TKI) and its derivatives, nilotinib and dasatinib, and the receptor-targeted TKIs gefinitib and erlotinib.Citation148 TKIs are most often prescribed for prolonged periods, often in combination with other drugs, including antifungal agents, thus, an understanding of their effects on fungal development is essential, as several protein kinase classes regulate key intracellular signals that impact fungal cell differentiation and death.Citation149

Effects on Candida and Aspergillus.

The only reported direct effects of TKIs on Candida and Aspergillus spp. are derived from in vitro observations of TKIs not in clinical use. Work involving the TKIs genistein, herbimycin, tyrphostin-47 and calphostin C provide insight into what may be expected for the modulation of key aspects of Candida biology by TKIs in general ().

Genistein appears to positively impact the growth of, rate of germination of and Hsp90 expression in vitro in C. albicans.Citation150 In view of its activity as a phytoestrogen, these effects may result from interaction of it with, for example, the Candida estrogen-binding protein. Both genistein and tyrphostin-47 have blocked Candida-induced tyrosine phosphorylation and subsequent endocytosis by endothelial cells.Citation151 C. albicans is known to induce its own uptake by endothelial cells, a process that requires tyrosine phosphorylation.Citation151 Using immunofluorescence microscopy, researchers observed that phosphotyrosine-containing endothelial cell proteins associated with the cell cytoskeleton surrounding C. albicans germ tubes while undergoing endocytosis.Citation151 Blocking of adhesion of endothelial cells to vitronectin molecules could result in inhibition of the critical first step in the escape of C. albicans from the intravascular compartment.

In Aspergillus spp., phagocytosis of conidia is an actindependent process that requires TK activity. In one study, two broad-range TKIs (genistein and AG18) and inhibitors that specifically blocked src kinases strongly reduced A. fumigatus conidia uptake by human and murine macrophages.Citation152 This could have implications for Aspergillus virulence as phagocytosis of conidia (e.g., in the lung) is required for their elimination.

Interactions with antifungal drugs. In addition to directly affecting fungi, TKIs are extensively metabolized by cytochrome P450 (CYP) enzymes.Citation148 Certain TKIs are also substrates or inhibitors of the drug transporters P-glycoprotein (Pgp), breast cancer resistance protein and organic cation carrier 1.Citation153 Because of their potential drug-drug interactions with azole antifungals, understanding the pharmacokinetics of TKIs is essential. Imatinib is metabolized primarily by the CYP isoenzyme 3A4 but is also a substrate of organic cation carrier 1, Pgp and BRCP. Fluconazole and itraconazole are dual CYP3A4 and Pgp inhibitors; thus, administration of them with imatinib may lead to increased plasma and intracellular concentrations of imatinib and increased toxicity of this agent.Citation148 Drug-drug interactions should be considered when administering TKIs with other inhibitors of the CYP3A family, most notably voriconazole. Similar interactions can be expected for dasatinib and nilotinib.Citation148

Autophagy Inhibitors

Autophagy is a conserved process by which eukaryotes recycle cytoplasm and organelles for survival during stress and starvation. Implicated in cancer development, autophagy is also harnessed by host cells to eliminate microorganisms.Citation154 It may be induced by rapamycin exposure via inhibition of TOR kinase and subsequent activation of the autophagy pathway.Citation154Citation156 Active autophagy is likely to be incompatible with rapid fungal growth.Citation155 Inhibition of the process of autophagy may occur by pharmacological means, such as class I and III inhibitors of phosphatidylinositol-3-kinase—3-methyladenine and wortmannin, respectively—and their derivativesCitation11 or by disruption of genes (i.e., ATG) encoding for autophagy proteins (e.g., Atg1 and Atg8 in S. cerevisiae).Citation156 Because the functional significance of autophagy in pathogenic fungi has yet to be established, the effects of autophagy inhibitors on fungi can only be surmised.

Effects on Candida.

Autophagy appears to have a minor role in C. albicans yeast-hyphal transition, chlamydospore differentiation and virulence in mice with systemic candidiasis as demonstrated in experiments using autophagy-defective mutants without the ATG9 gene homolog.Citation157 However, autophagy-defective C. glabrata mutants lacking expression of the CgATG11 and CgATG17 genes were associated with decreased survival in C. glabrata, with the double mutant being highly sensitive to engulfment by macrophages.Citation158 Current data suggest that autophagy inhibitors (as well as other agents that indirectly induce autophagy) reduce survival of C. glabrata in vitro but have little direct effect on C. albicans survival and morphogenesis. Yet since autophagy is profoundly important to eukaryote cell biology, it is unlikely to be completely superfluous in C. albicans.

Effects on Aspergillus.

Reported data on the direct effects of autophagy inhibitors on Aspergillus development are also scarce, although Aspergillus spp. are reported to be resistant to wortmannin.Citation159 In Aspergillus oryzae autophagy appears to be involved in nutrient recycling during starvation; mutants of this species with deletions of the ATG1 or ATG8 gene were unable to undergo asexual development and acquire nutrients.Citation159,Citation160 In a study of A. fumigatus, autophagy was required for aerial hyphae formation, conidial germination, hyphal foraging and growth under nutrient-deficient conditions; recent data have also suggested that autophagy contributes to metal ion homeostasis but the mechanism by which this occurs requires further investigation.Citation155,Citation161 As for C. albicans, autophagy does not appear to be essential for virulence in mice with aspergillosis,Citation155,Citation161 the reason for which remains to be defined. One possible explanation is that for A. fumigatus, the fungus is able to survive on dead/decaying matter and is relatively resistant to nutrient stress.Citation152

Perspectives

Collectively, the scientific literature indicates that antineoplastic and other immunosuppressive therapies can directly affect fungal growth, development and virulence. Certain classes of anticancer agents, such as the calcineurin inhibitors and HDACis may significantly influence antifungal drug susceptibility. The specific effects and magnitude thereof of these agents on each of these aspects of fungal biology vary both with the agent class and within each class. Of note, these effects are observed not only with new antineoplastic agents but also with established cytotoxic drugs.

Studies of novel and investigational anticancer drugs show that signal transduction networks that modulate crucial events during fungal invasion can be targeted in the development of drug-pathogen interaction-based cancer-management strategies. Pertinent observations in these studies include findings that inhibitors of calcineurin and/or TOR perturb morphogenesis, membrane stress responses, survival of fungi in serum and fungal virulence in experimental models of infection. These inhibitors can also reduce azole tolerance and may be considered for novel therapies for recalcitrant azole-resistant candidiasis and biofilmassociated infections. Results in Aspergillus suggest that these inhibitors inhibit growth and filamentation, but in vivo animal data are conflicting. Inhibition of candidal Hsp90 activity and modulation of fungal gene expression by Candida-specific HDAC inhibition indicate that they are feasible approaches to the treatment of invasive candidiasis. TKIs have good potential for inhibiting fungal entry into epithelial cells and subsequent phagocytosis by macrophages but may interact significantly with antifungal agents.

These data however, should be considered in the context in which they were obtained. Specifically, researchers largely observed the direct effects of all classes of anticancer agents on Candida and Aspergillus spp. under in vitro conditions. Uncertainty regarding the wider applicability of these results and their clinical relevance stems from the use of these “artificial” laboratory conditions, incorporating technical variables such as the effects of different media, inocula and growth conditions. Furthermore, investigators conducted many of the experiments using only single strains or small numbers of strains. Testing of larger numbers of strains of given species is required to determine drug effects in a population-based context and confirm prior observations. It is intriguing that the use of anticancer/immunosuppressive agents with the potential to affect fungal biology or growth largely occurs is in a clinical context where there is a relatively high rate of IFIs (i.e., in transplant and cancer patients). These “paradoxical” observations emphasize the complexicty of the host immune response and host-pathogen interactions and that laboratory observations, even if robust, should not be used alone to predict clinical response to a drug.

Another limitation of the current data set is evaluation of antineoplastic drugs as single agents. Because these drugs are typically given in combinations, assessment of their net combined effects on fungal biology is required. To date, these combined effects remain unknown. Anticancer agents that may have no discernible effects when given alone may well have significant effects when given with other agents. Further clinical studies, in particular those testing the in vivo synergy of antineoplastic agents with currently used antifungals, are warranted.

Conclusions

In summary, the direct effects of antineoplastic agents on the development of Candida and Aspergillus infections and the virulence of their species as well as on antifungal drug resistance are varied. Many questions remain unanswered. In particular, results obtained using both in vitro and in vivo experimental systems may not reflect the clinical context in which a host is exposed to a defined inoculum of a fungus in a complex, pleiotropic environment (). The resultant ability of various antineoplastic and immunosuppressive agents to create permissive conditions for the iniation and progression of the fungal are multifaceted and difficult to predict. The occurrence of such infection is dependent on not only the intrinsic virulence properties of the fungal strain, but also local and systemic metabolic factors and the effects of antifungal and other chemotherapeutic agents in the pathogen's environmental milieu. Because fungal infections frequently begin on mucosal surfaces in multi-organism communities, chemotherapeutic drugs may also affect the quorum-sensing ability of the infecting organisms. Evaluation of the effects on fungi of antineoplastic drugs administered with and without antifungal agents in more clinically plausible systems that reflect “real-life” scenarios is awaited with interest.

Abbreviations

5AC=

5-azacytidine

17-AAG=

17-allylamino-17-demethoxygeldanamycin

ara-C=

cytosine arabinoside

AMB=

amphotericin B

ATO=

arsenic trioxide

CDR =

Candida drug resistance

Crz1p=

calcineurin-regulated zinc finger-containing 1 protein

CsA=

cyclosporine

17,-DMAG=

17-desmethoxy-17-N,N-diemthylaminoethylaminogeldanamycin

FK-506=

tacrolimus

FKBP12=

tacrolimus binding protein 12

5FU=

5-fluorouracil

Hsp90=

heat shock protein 90

HDAC=

histone deacytelase

HDACi=

histone deacetylase inhibitor

IFI=

invasive fungal infection

MDR =

multidrug resistance

MG-CD0103=

mocetinostat

MIC=

minimal inhibitory concentration

PG-paclitaxel=

polyglutamate bound paclitaxel

Pgp=

P-glycoprotein

SRE=

steroid responsive element

TSA=

trichostatin A

SAHA=

suberoylanilide hydroxamic acid

TK=

tyrosine kinase

TKI=

tyrosine kinase inhibitor

TOR=

target of rapamycin

TORC=

target of rapamycin complex

Figures and Tables

Figure 1 Mammalian targets of cancer chemotherapy agents reported to affect the biology of pathogenic fungi. Arrows represents activation; blocked lines represent inhibition. NFATc, nuclear factor of activated T cells; HSP90, heat shock protein 90; HSP70, heat shock protein 70; P, Phosphate group.

Figure 1 Mammalian targets of cancer chemotherapy agents reported to affect the biology of pathogenic fungi. Arrows represents activation; blocked lines represent inhibition. NFATc, nuclear factor of activated T cells; HSP90, heat shock protein 90; HSP70, heat shock protein 70; P, Phosphate group.

Table 1 Direct effects of antineoplastic agents and antibiotics on the growth and virulence of Candida and Aspergillus

Table 2 Direct effects of corticosteroids and other immunosuppressive agents used in transplantation on the growth and virulence of Candida and Aspergillus spp.

Acknowledgments

This research is supported in part by the National Institutes of Health through MD Anderson's Cancer Center Support Grant CA016672.

References

  • Pfaller MA, Diekema DJ. Epidemiology of invasive mycoses in North America. Crit Rev Microbiol 2010; 36:1 - 53
  • Fishman JA. Infection in solid organ transplant recipients. N Engl J Med 2007; 357:601 - 614
  • Pappas PG, Alexander BD, Andes DR, Hadley S, Kauffman CA, Freifeld A, et al. Invasive fungal infections among organ transplant recipients: results of the transplant-associated infection surveillance network (TRANSNET). Clin Infect Dis 2010; 50:101 - 111
  • Marr KA, Carter RA, Boeckh M, Martin P, Corey L. Invasive aspergillosis in allogeneic stem cell transplant recipients: changes in epidemiology and risk factors. Blood 2002; 100:4358 - 4366
  • Lionakis MS, Kontoyiannis DP. Glucocorticoids and invasive fungal infections. Lancet 2003; 362:1828 - 1838
  • Rubin RH. Overview: pathogenesis of fungal infections in the organ transplant recipient. Transpl Infect Dis 2002; 4:12 - 17
  • MacFadden KD, Saito S, Pruzanski W. The effect of chemotherapeutic agents on chemotaxis and random migration of human leukocytes. J Clin Oncol 1985; 3:415 - 419
  • Ng TT, Robson GD, Denning DW. Hydrocortisone-enhanced growth of Aspergillus spp.: implications for pathogenesis. Microbiology 1994; 140:2475 - 2479
  • Ueta E, Tanida T, Yoneda K, Yamamoto T, Osaki T. Increase of Candida cell virulence by anticancer drugs and irradiation. Oral Microbiol Immunol 2001; 16:243 - 249
  • Bastidas RJ, Reedy JL, Morales-Johansson H, Heitman J, Cardenas ME. Signalling cascades as drug targets in model and pathogenic fungi. Curr Opin Invest Drugs 2008; 9:856 - 864
  • Cardenas ME, Cruz MC, Del Poeta M, Chung N, Perfect JR, Heitman J. Antifungal activities of antineoplastic agents: Saccharomyces cerevisiae as a model system to study drug action. Clin Microbiol Rev 1999; 12:583 - 611
  • Swain SM. Chemotherapy: updates and new perspectives. Oncologist 2010; 15:8 - 17
  • Sharp S, Workman P. Inhibitors of the HSP90 molecular chaperone: current status. Adv Cancer Res 2006; 95:323 - 348
  • Rotili D, Simonetti G, Savarino A, Palamara AT, Migliaccio AR, Mai A. Non-cancer uses of histone deacetylase inhibitors: effects on infectious diseases and β-hemoglobinopathies. Curr Top Med Chem 2009; 9:272 - 291
  • Kufe DW, Major PP, Egan EM, Beardsley GP. Correlation of cytotoxicity with incorporation of ara-C into DNA. J Biol Chem 1980; 255:8997 - 9000
  • Perez EA. Microtubule inhibitors: Differentiating tubulin-inhibiting agents based on mechanisms of action, clinical activity and resistance. Mol Cancer Ther 2009; 8:2086 - 2095
  • Ghannoum MA, Abu-Elteen KH, Motawy MS, Abu-Hatah MA, Ibrahim AS, Criddle RS. Combinations of antifungal and antineoplstic drugs with interactive effects of inhibition of yeast growth. Chemotherapy 1990; 36:308 - 320
  • Savani DV, Perfect JR, Durack DT. Activity of anticancer drugs against yeast in vitro. Program and abstracts of the 26th Interscience Conference on Antimicrobial Agents and Chemotherapy 1986; 838:Washington DC American Society for Microbiology 252
  • Graybill JR, Bocanegra R, Fothergill A, Rinaldi MG. Bleomycin therapy of experimental disseminated candidiasis in mice. Antimicrob Agents Chemother 1996; 40:816 - 818
  • Bleomycin http://www.drugs.com/pro/bleomycin.html
  • Ghannoum MA, Motawy MS, Abu-Hatah MA, Abu-Elteen KA, Criddle RS. Interactive effect of antifungal and antineoplastic agents on yeasts commonly prevalent in cancer patients. Antimicrob Agents Chemother 1989; 33:726 - 730
  • Ghannoum MA, Al-Kharis A. Effect of antineoplastic agents on the growth and ultrastructure of Candida albicans. Mykosen 1984; 27:452 - 464
  • Land GA, Hulme KL, Chaffin WL. The effect of selected antineoplastic agents on the morphology of Candida albicans 5865. Can J Microbiol 1980; 26:812 - 818
  • Linaris CEB, Briebeler D, Cargnelutti D, Alves SH, Morsch VM, Schetinger MRC. Catalase activity in Candida albicans exposed to antineoplastic drugs. J Med Microbiol 2006; 55:259 - 262
  • Nakagawa Y, Kanbe T, Mizuguchi I. Disruption of the human pathogenic yeast Candida albicans catalase gene decreased survival in mouse-model infection and elevated susceptibility to higher temperature and to detergents. Microbiol Immunol 2003; 47:395 - 403
  • Ghannoum MA. Effects of antineoplastic agents on growth, morphology and metabolism of Torulopsis glabrata. Mycopathologica 1986; 95:171 - 181
  • Miyamoto CT, Sant'Anna JR, Franco CCS, Castro-Prado MAA. Genotoxicity (mitotic recombination) of the cancer chemotherapeutic agents cisplatin and cytosine arabinoside in Aspergillus nidulans. Food Chem Toxicol 2007; 45:1091 - 1095
  • Becker TCA, Castro-Prado MAA. Parameiosis in Aspergillus nidulans in response to doxorubicin. Folia Microbiol 2004; 49:699 - 704
  • Tamame M, Antoquera F, Santos E. Developmental characterisation and chromosomal mapping of the 5-azacytidine-sensitive fluF locus of Aspergillus nidulans. Mol Cell Biol 1988; 8:3043 - 3050
  • Ben-Ami R, Varga J, Lewis RE, May GS, Nierman WC, Kontoyiannis DP. Characterization of a 5-azacytidine-induced developmental Aspergillus fumigatus variant. Virulence 2010; 1:164 - 173
  • Pharmion Corporation. Azacitidine Investigator's brochure 2007; Boulder, CO 5
  • Moussa NM, Ghannoum MA, Whittaker PA, el-Ezaby MS, Quraman S. Effects of cisplatin and tow novel palladium complexes on Candida albicans. Microbios 1990; 62:252 - 253
  • Chandrasekar K, Shyla JH, Malathi R. Can antitumor platinum compounds be effective against Candida albicans?—a screening assay fusing disk diffusion method. J Clin Microbiol 2000; 38:3905
  • Salas S, Mercier C, Ciccolini J, Pourroy B, Fanciullino R, Tranchand B, et al. Therapeutic drug monitoring for dose individualization of Cisplatin in testicular cancer patients based upon total platinum measurement in plasma. Ther Drug Monit 2006; 28:532 - 539
  • Malathi R, Chandrasekar K. Self splicing in group-I intron of Tetrahymena rRNA and its possible inhibition when reacted with platinum complexes. J Biosci 1999; 24:96
  • Sarachek A, Henderson LA. Modification of responses of Candida albicans to cisplatin by membrane damaging antimycotic agents. Mycoses 1991; 34:177 - 182
  • Chandrasekar K, Raghunathan M, Ganesan N. Morphological and growth altering effects of Cisplatin in C. albicans using fluorescence microscopy. Ann Clin Microbiol Antimicrob 2005; 4:7
  • Litzow MR. Arsenic trioxide. Expert Opin Pharmacother 2008; 9:1773 - 1785
  • Jin YH, Dunlop PE, McBride SJ, Al-Refai H, Bushel PR, Freedman JH. Global transcriptome and deletome profiles of yeast exposed to transition metals. PLoS Genet 2008; 4:1000053
  • Hosiner D, Lempiainen, Reiter W, Urban J, Loewith R, Ammerer G, et al. Arsenic toxicity to Saccharomyces cerevisiae is a consequence of inhibition of the TORC1 kinase combined with a chronic stress response. Mol Biol Cell 2009; 20:1048 - 1057
  • Buckova M, Godocikova J, Polek B. Responses in the mycelia growth of Aspergillus niger isolates to arsenic contaminated environments and their resistance to exogenic metal stress. J Basic Microbiol 2007; 47:295 - 300
  • Buckova M, Godocikova J, Simonovicova A, Polek B. Production of catalases by Aspergillus niger isolates as a response to pollutant stress by heavy metals. Curr Microbiol 2005; 50:175 - 179
  • Zhao J, Kim JE, Reed E, Li QQ. Molecular mechanism of antitumour activity of taxanes in lung cancer. Int J Oncol 2005; 27:247 - 256
  • Whelan J. Targeted taxane therapy for cancer. Drug Discov Today 2002; 7:90 - 92
  • Chakravarthi BVSK, Das P, Surendranath K, Karande AA, Jayabaskaran C. Production of paclitaxel by Fusarium solani isolated from Taxus celebica. J Biosci 2008; 33:259 - 267
  • Foland TB, Dentler WL, Suprenant KA, Gupta ML Jr, Himes RH. Paclitaxel-induced microtubule stabilisation causes mitotic block and apoptotic-like cell death in a paclitaxel-sensitive strain of Saccharomyces cerevisiae. Yeast 2005; 22:971 - 978
  • Yokoyama K, Kaji H, Nishimura K, Miyaji M. The role of microfilaments and microtubules in apical growth and dimorphism of Candida albicans. J Gen Microbiol 1990; 136:1067 - 1075
  • Filler SG, Swerdloff JN, Hobbs C, Luckett PM. Penetration and damage of endothelial cells by Candida albicans. Infect Immun 1995; 63:976 - 983
  • Maraki S, Hajiioannou I, Anatoliotakis N, Plataki M, Chatzinikolaou I, Zoras O, et al. Ceftriaxone and dexamethasone affecting yeast gut flora in experimental mice. J Chemother 1999; 11:363 - 366
  • Myerowitz RL. Gastrointestinal and disseminated candidiasis. An experimental model in the immunosuppressed rat. Arch Pathol Lab Med 1981; 105:138 - 143
  • Ellepola ANB, Samaranayake LP. Inhalational and topical steroids, and oral candidosis: a mini-review. Oral Dis 2001; 7:211 - 216
  • Bykov VL. Morphological analysis of the effect of corticosteroids on the development and course of vaginal candidiasis. Akush Ginekol 1989; 2:35 - 37
  • Ghannoum MA, Elteen KA. Effect of growth of Candida spp. in the presence of various glucocorticoids on the adherence to human buccal epithelial cells. Mycopathologia 1987; 98:171 - 178
  • Wu TG, Wilhelmus KR, Mitchell BM. Experimental keratomycosis in a mouse model. Invest Ophthalmol Vis Sci 2003; 44:210 - 216
  • Gyetvai A, Emri T, Fekete A, Varga Z, Gazdag Z, Pesti M, et al. High dose methylprednisolone influences the physiology and virulence of Candida albicans ambiguously and enhances the candidacidal activity of the polyene antibiotic amphotericin B and the superoxide-generating agent menadione. FEMS Yeast Res 2007; 7:265 - 275
  • Cutler JE. Putative virulence factors of Candida albicans. Annu Rev Microbiol 1991; 45:187 - 218
  • Loose DS, Schurman DJ, Feldman D. A corticosteroid binding protein and endogenous ligand in Candida albicans indicating a possible steroid-receptor system. Nature 1981; 293:477 - 479
  • Stover EP, Loose DS, Stevens DA, Feldman D. Ketoconazole binds to the intracellular corticosteroid-binding protein in Candida albicans. Biochim Res Commun 1983; 117:43 - 50
  • Banerjee D, Martin N, Nandi S, Shukla S, Dominguez A, Mukhopadhyay G, et al. Genome-wide steroid response study of the major human fungal pathogen Candida albicans. Mycopathologia 2007; 164:1 - 17
  • Karnani N, Gaur NA, Jha S, Puri N, Krishnamurthy S, Goswami SK, et al. SRE1 and SRE2 are two specific steroid-responsive modules of Candida drug resistance gene 1 (CDR1) promoter. Yeast 2004; 21:219 - 239
  • Ramondec I, Pinel C, Ambroise-thomas P, Grillot R. Does hydrocortisone modify the in vitro susceptibility of Aspergillus fumigatus to itraconazole and amphotericin B?. Med Mycol 1998; 36:69 - 73
  • Steinbach WJ, Reedy JL, Cramer RA Jr, Perfect JR, Heitman J. Harnessing calcineurin as a novel anti-infective agent against invasive fungal infections. Nat Rev Microbiol 2007; 5:418 - 430
  • Fox DS, Heitman J. Good fungi gone bad: the corruption of calcineurin. BioEssays 2002; 24:893 - 903
  • Heitman J, Movva NR, Hall MN. Targets for cell cycle arrest by the immunosuppressant rapamycin in yeast. Science 1991; 253:905 - 909
  • Cutler NS, Pan X, Heitman J, Cardenas ME. The TOR signal transduction cascade controls cellular differentiation in response to nutrients. Mol Cell Biol 2001; 12:4103 - 4113
  • High KP. The antimicrobial activities of cyclosporine, FK506 and rapamycin. Transplantation 1994; 12:1689 - 1700
  • Vakil R, Knilans K, Andes D, Kwon GS. Combination antifungal therapy involving amphotericin B, rapamycin and 5-FC using PEG-phospholipid micelles. Pharm Res 2008; 25:2056 - 2064
  • Cruz MC, Goldstein AL, Blankenship J, Del Poeta M, Perfect JR, McCusker JH, et al. Rapamycin and less immunosuppressive analogs are toxic to Candida albicans and Cryptococcus neoformans via FKBP12-dependent inhibition of TOR. Antimicrob Agents Chemother 2001; 45:3162 - 3170
  • Liao WL, Ramon AM, Fonzi WA. GLN3 encodes a global regulator of nitrogen metabolism and virulence of Candida albicans. Fungal Genet Biol 2008; 45:514 - 526
  • Baker H, Sidorowicz A, Sehgal SN, Vezina C. Rapamycin (AY-22,989), a new antifungal antibiotic: III. In vitro and in vivo evaluation. J Antibiot 1978; 31:539 - 545
  • Onyewu C, Blankenship JR, Del Poeta M, Heitman. Ergosterol biosynthesis become fungicidal when combined with calcineurin inhibitors against Candida albicans, Candida glabrata and Candida krusei. Antimicrob Agents Chemother 2003; 47:956 - 964
  • Marchetti O, Moreillon P, Glauser MP, Bille J, Sanglard D. Potent synergism of the combination of fluconazole and cyclosporine in Candida albicans. Antimicrob Agents Chemother 2000; 44:2932 - 2938
  • Cruz M, Goldstein AL, Blankenship JR, Del Poeta M, Davis D, Cardenas ME, et al. Calcineurin is essential for survival during membrane stress in Candida albicans. EMBO J 2002; 21:546 - 559
  • Bader T, Bodendorfer B, Schroppel K, Morschhauser J. Calcineurin is essential for virulence in Candida albicans. Infect Immun 2003; 71:5344 - 5354
  • Blankenship JR, Wormley FL, Boyce MK, Schell WA, Filler SG, Perfect JR, et al. Calcineurin is essential for Candida albicans survival in serum and virulence. Euk Cell 2003; 2:422 - 430
  • Onyewu C, Wormley FL Jr, Perfect JR, Heitman J. The calcineurin target Crz1, functions in azole tolerance but is not required for virulence of Candida albicans. Infect Immun 2004; 72:7330 - 7333
  • Marchetti O, Entenza JM, Sanglard D, Bille J, Glauser MP, Moreillon P. Fluconazole plus cyclosporine: a fungicidal combination effective against experimental endocarditis due to Candida albicans. Antimicrob Agents Chemother 2000; 44:2932 - 2938
  • Reedy JL, Husain S, Ison M, Pruett TL, Singh N, Hietman J. Immunotherapy with tacrolimus (FK506) does not select for resistance to calcineurin inhibitors in Candida albicans isolates from liver transplant patients. Antimicrob Agents Chemother 2006; 50:1573 - 1577
  • Onyewu C, Ahshari N, Heitman J. Calcineurin promotes infection of the cornea by Candida albicans. Antimicrob Agents Chemother 2006; 50:3963 - 3965
  • Uppuluri P, Nett J, Heitman J, Andes D. Synergistic effect of calcineurin inhibitors and fluconazole against Candida albicans biofilms. Antimicrob Agents Chemother 2008; 52:1127 - 1132
  • Miyazaki T, Yamauchi S, Inaminbe T, Nagayoshi Y, Saijo T, Izumikawa K, et al. Roles of calcineurin and Crz1 in antifungal susceptibility and virulence of Candida glabrata. Antimicrob Agents Chemother 2010; 54:1639 - 1643
  • Palmbad J, Lonnqvist B, Carlsson B, Grimfors G, Jarnmark M, Lerner R. Oral ketoconazole prophylaxis for Candida infections during induction therapy for acute leukemia in adults: more bacteremia. J Int Medicine 1992; 231:363 - 370
  • Kern W, Behre G, Rudolf T. failure of fluconazole prophylaxis to reduce mortality or the requirement of systemic amphtoericin B therapy during treatment for refractory AML: results of a prospective randomised phase III study. German AML Cooperative Group. Cancer 1998; 83:291 - 301
  • Krajewska-Kulak E, Niczyporuk W. J Effects of cyclosporine A on mycelia transformation of Candida albicans in human serum. Eur Acad Dematol Venereol 1998; 10:191 - 192
  • Steinbach WJ, Cramer RJ Jr, Perfect BZ, Asfaw YG, Sauer TC, Najvar LK, et al. Calcineurin controls growth, morphology and pathogencity in Aspergillus fumigatus. Eukayrot Cell 2006; 5:1091 - 1103
  • Ferreira ME, Heinekamp T, Hartl A, Brakhage AA, Semighini CP, Harris SD, et al. Functional characterisation of the Aspergillus fumigatus calcineurin. Fungal Genet Biol 2007; 44:219 - 230
  • Steinbach WJ, Singh N, Miller JL, Benjamin DK, Schell WA, Heitman J, et al. In vitro interaction between antifungals and immunosuppressants against Aspergillus fumigatus isolates from transplant and nontransplant patients. Antimicrob Agents Chemother 2004; 48:4922 - 4925
  • Steinbach WJ, Schell WA, Blankenship JR, Onyewu C, Heitman J, Perfect JR. In vitro interactions between antifungals and immunosuppressants against Aspergillus fumigatus. Antimicrob Agents Chemother 2004; 48:1664 - 1669
  • Kontoyiannis DP, Lewis RE, Osherov N, Albert ND, May GS. Combination of caspofungin with inhibitors of the calcineurin pathway attenuates growth in vitro in Aspergillus species. J Antimicrob Chemother 2003; 51:313 - 316
  • Berenguer J, Allende MC, Lee JW, Garrett K, Lyman C, Ali NM, et al. Pathogenesis of pulmonary aspergillosis. Granulocytopenia versus cyclosporine and methylpredisolone-induced immunosuppression. Am J Resp Crit Care 1995; 152:1079 - 1086
  • High KP, Washburn RG. Invasive aspergillosis in mice immunosuppressed with cyclosporine A, tacrolimus (FK506) or sirolimus (rapamycin). J Infect Dis 1997; 175:222 - 225
  • Nielsen Kahn J, Hsu MJ, Racine F, Giacobbe R, Motyl M. Caspofungin susceptibility in Aspergillus and non-Aspergillus moulds: inhibition of glucan synthase and reduction of B-D-1,3 glucan levels in culture. Antimicrob Agents Chemother 2006; 50:2214 - 2216
  • Steinbach WJ, Cramer RA, Perfect BZ, Henn C, Nielsen K, Heitman J, et al. Calcienurin inhibition or mutation enhances cell wall inhibitors against Aspergillus fumigatus. Antimicrob Agents Chemother 2007; 51:2979 - 2981
  • Cramer RA Jr, Perfect BZ, Pinchai N, Park S, Perlin DS, Asfaw YG, et al. Calcineurin target CrzA regulates conidial germination, hyphal growth and pathogenesis of Aspergillus fumigatus. Eukaryot Cell 2008; 7:1085 - 1097
  • Cruz MC, Del Poeta M, Wang P, Wenger R, Zenke G, Quesniaux VF, et al. Immunosuppressive and nonimmunosuppressive cyclosporine analogs are toxic to the opportunistic fungal pathogen Cryptococcus neoformans via cyclophilin-dependent inhibition of calcineurin. Antimicrob Agents Chemother 2000; 44:143 - 149
  • Taldone T, Gozman A, Maharaj R, Chiosis G. Targeting Hsp90: small-molecule inhibitors and their clinical development. Curr Opin Pharmacol 2008; 8:370 - 374
  • Donnelly A, Blagg BSJ. Novobiocin and additional inhibitors of the Hsp90 C-terminal nucleotide-binding pocket. Curr Med Chem 2008; 15:2702 - 2717
  • Kamal A, Boehm M, Burrows FJ. Therapeutic and diagnostic implications of Hsp90 activation. Trends Mol Med 2004; 10:283 - 290
  • Cowen LE, Lindquist S. Hsp90 potentiates the rapid evolution of new traits: drug resistance in diverse fungi. Science 2005; 309:2185 - 2189
  • Cowen LE, Carpenter AE, Matangkasombuk O, Fink GR, Lindquist S. Genetic architecture of Hsp90-dependent drug resistance. Euk Cell 2006; 5:2184 - 2188
  • Cowen L, Singh SD, Kohler JR, Collins C, Zaas AK, Schell WA, et al. Harnessing Hsp90 function as a powerful, broadly effective therapeutic strategy for fungal infectious disease. PNAS 2009; 106:2818 - 2823
  • Singh SD, Robbins N, Zaas AK, Schell WA, Perfect JR, Cowen LE. Hsp90 governs echinocandin resistance in the pathogenic yeast Candida albicans via calcineurin. PLoS Pathogens 2009; 5:1000532
  • Kaneko Y, Ohno H, Imamura Y, Kohno S, Miyazaki Y. The effects of an Hsp90 inhibitor on the paradoxical effect. Jpn J Infect Dis 2009; 62:392 - 393
  • Shapiro RS, Cowen LE. Coupling temperature sensing and development. Virulence 2010; 1:45 - 48
  • Hodgetts S, Nooney L, Al-Akeel R, Curry A, Awad S, Matthews R, et al. Efungumab and caspofungin: pre-clinical data supporting synergy. J Antimicrob Chemother 2008; 61:1132 - 1139
  • Matthews RC, Burnie JP. Human recombinant antibody to HSP90: a natural partner in combination therapy. Curr Mol Med 2005; 5:403 - 411
  • Pachl J, Svoboda P, Jacobs F, Vandewoude K, Hoven B, van der Spronik P, et al. A randomised blinded multicentre trial of lipid-associated amphotericin B alone versus in combination with an antibody-based inhibitor of heat shock protein 90 in patients with invasive candidiasis. Clin Infect Dis 2006; 42:1404 - 1413
  • Srivastava P. Roles of hear shock proteins in innate and adaptive immunity. Nat Rev Immunol 2002; 2:185 - 194
  • Andrews KT, Tan TN, Lucke AJ, Kahnberg P, Lee GT, Boyle GM, et al. Potent antimalarial activity of histone acetylase inhibitor analogues. Antimicrob Agents Chemother 2008; 52:1454 - 1461
  • Batty N, Malouf GG, Issa JP. Histone deacetylase inhibitors as anti-neoplastic agents. Cancer Lett 2009; 280:192 - 200
  • Copeland A, Buglio D, Younes A. Histone deacetylase inhibitors in lymphoma. Curr Opin Oncol 2010; 22:431 - 436
  • Brosch G, Loidl P, Graessle S. Histone modifications and chromatin dynamics: a focus on filamentous fungi. FEMS Microbiol Rev 2008; 32:409 - 439
  • Wurtele H, Tsao S, Lepine G, Mullick A, Tremblay J, Drogaris P, et al. Modulation of histone H3 lysine 56 acetylation as an antifungal therapeutic strategy. Nat Med 2010; 16:774 - 781
  • Srikantha T, Tsai L, Daniels K, Klar AJS, Soll DR. The histone deacetylase genes HDA1 and RPD3 play distinct roles in regulation of high frequency phenotypic switching in Candida albicans. J Bacteriol 2001; 183:4614 - 4625
  • Smith WL, Edlind TD. Histone deacetylase inhibitors enhance Candida albicans sensitivity to azoles and related antifungals: correlation with reduction in CDR and ERG upregulation. Antimicrob Agents Chemother 2002; 46:3532 - 3539
  • Simonetti G, Passariello C, Rotili D, Mai A, Garaci E, Palamara AT. Histone deacetylase inhibitors may reduce pathogenicity and virulence in Candida albicans. FEMS Yeast Res 2007; 7:1371 - 1380
  • Mai A, Rotili D, Massa S, Brosch G, Simonettti G, Passariello C, Palamara AT. Discovery of uracil-based histone deacetylase inhibitors able to reduce acquired antifungal drug resistance and trailing growth in Candida albicans. Bioorg Med Chem 2007; 17:1221 - 1225
  • Kozikowski AP, Tapadar S, Luchini DN, Kim KH, Billadeau DD. Use of the nitrile oxide cycloaddition (NOC) reaction for molecular rpobe generation: a new class of enzyme selective histone decetylase inhibitors (HDACIs) showing picomolar activity at HDAC6. J Med Chem 2008; 51:4370 - 4373
  • Aoyagi S, Archer TK. Modulating molecular chaperone Hsp90 functions through reversible acetylation. Trends Cell Biol 2005; 15:565 - 567
  • da Rosa JL, Boyartchuk VL, Zhu LJ, Kaufman PD. Histone acertyltransferase Rtt109 is required for Candida albicans pathogenesis. Proc Natl Acad Sci 2010; 107:1594 - 1599
  • Pfaller MA, Messer SA, Georgopapadakou N, Martell LA, Besterman JM, Diekema DJ. Activity of MGCD290, a Hos histone deacetylase inhibtor in combination with azole antifungals against opportunistic fungal pathogens. J Clin Microbiol 2009; 47:3797 - 3804
  • Lee I, Oh JH, Shwab EK, Degenais T, Andes D, Keller NP. HdaA, a class 2 histone deacetylase of Aspergillus fumigatus, affects germination and secondary metabolite production. Fungal Genet Biol 2009; 46:782 - 790
  • Tribus M, Bauer I, Galehr J, Rieser G, Trojer P, Brosch G, et al. A novel motif in fungal class 1 histone deacetylases is essential for growth and development of Aspergillus. Mol Biol Cell 2010; 21:345 - 353
  • Shen LL, Fostel JM. DNA topoisomerase inhibitors as antifungal agents. Adv Pharmacol 1994; 29:227 - 244
  • Ozdek SC, Miller D, Flynn PM, Flynn HW Jr. In vitro antifungal activity of the fourth generation fluoroquinolones against Candida isolates from human ocular infections. Ocul Immunol Inflamm 2006; 14:347 - 351
  • Stergiopoulou T, Meletiadis J, Sein T, Papaioannidou P, Tsiouris I, Roilides E, et al. Isobolographic analysis of pharmacodynamic interactions between antifungal agents and ciprofloxacin against Candida albicans and Aspergillus fumigatus. Antimicrob Agents Chemother 2008; 52:2196 - 2204
  • Stergiopoulou T, Meletiadis J, Sein T, Papaioannidou P, Tsiouris I, Roilides E, et al. Comparative pharmacodynamic interaction analysis between ciprofloxacin, moxifloxacin and levofloxacin and antifungal agents against Candida albicans and Aspergillus fumigatus. J Antimicrob Chemother 2009; 63:343 - 348
  • Maurer N, Wong KF, Hope MJ, Cullis PR. Anomalous solubility behaviour of the antibiotic ciprofloxacin encapsulated in liposomes; a 1H-NMR study. Biophys Biochim Acta 1998; 1374:9 - 20
  • Nakajima R, Kitamura A, Someya K, Tanaka M, Sato K. In vitro and in vivo antifungal activities of DU-6859a, a fluoroquinolone, in combination with amphotericin B and fluconazole against pathogenic fungi. Antimicrob Agents Chemother 1995; 39:1517 - 1521
  • Sugar AM, Liu XP, Chen RJ. Effectiveness of quinolone antibiotics in modulating the effects of antifungal drugs. Antimicrob Agents Chemother 1997; 41:2518 - 2521
  • Deren YT, Ozdek S, Kalkanci A, Akyurek N, Hasanreisoglu B. Comparison of antifungal efficacies of moxifloxacin, liposomal amphotericin B and combination treatment in experimental Candida albicans endophthalmitis in rabbits. Can J Microbiol 2010; 56:1 - 7
  • Shalit I, Horev-Azaria L, Fabian I, Blau H, Kariv N, Shechtman I, et al. Immunomodulatory and protective effects of moxifloxacin against Candida albicans-induced bronchopneumonia in mice injected with cyclophosphamide. Antimicrob Agents Chemother 2002; 46:2442 - 2449
  • Parker MA, King V, Howard KP. Nuclear magnetic resonance study of doxorubicin binding to cardiolipin containing magnetically orientated phospholipid bilayers. Biochim Biophys Acta Biomembranes 2001; 1514:206 - 216
  • Peiris V, Oppenheim BA. Antimicrobial activity of cytotoxic drugs may influence isolation of bacteria and fungi from blood cultures. J Clin Pathol 1993; 46:1124 - 1125
  • Kinnunen U, Syrjala H, Koistinen P, Koskela M. Idarubicin inhibits the growth of bacteria and yeasts in an automated blood culture system. Eur J Clin Microbiol Infect Dis 2009; 28:301 - 303
  • Kwok SC, Schelenz S, Wand X, Steverding D. In vitro effect of DNA topoisomerase inhibitors on Candida albicans. Med Mycol 2010; 48:155 - 160
  • O'Keefe J, Doyle S, Kavanagh K. Exposure of the yeast Candida albicans to the anti-neoplastic agent adriamycin increases the tolerance to amphotericin B. J Pharm Pharmcol 2003; 5:1629 - 1633
  • Kofla G, Turner V, Schulz B, Storch U, Froelich D, Rognon B, et al. Doxorubicin induces drug efflux pumps in Candida albicans. Med Mycol 2011; 49:132 - 142
  • Becker TCA, De Castro-Prado MAA. Parameiosis in Aspergillus nidulans in response to doxorubicin. Folia Microbiol 2004; 49:699 - 704
  • Jayaguru P, Raghunathan M. Group I intron renders differential susceptibility of Candida albicans to bleomycin. Mol Biol Rep 2007; 34:11 - 17
  • Kafer E. Botran and bleomycin induce crossing-over and bleomycin also increases aneuploidy in diploid strains of Aspergillus. Mutat Res 1990; 241:49 - 66
  • Moore CW, McKoy J, Del Valle R, Armstrong D, Bernard EM, Katz N, et al. Fungal cell wall septation and cytokinesis are inhibited by bleomycins. Antimicrob Agents Chemother 2003; 47:3281 - 3289
  • Alfeltra J, Verwiej PE. Antifungal activity of nonantifungal drugs. Eur J Clin Microbiol Infect Dis 2003; 22:397 - 407
  • Beggs WH. Combined activity of ketoconazole and sulphamethoxazole against Candida albicans. J Antimicrob Chemother 1982; 10:539 - 541
  • Chang MR, Cury AE. Amphotericin B-metronidazole combination against Candida spp. Rev Iberoam Micol 1998; 15:78 - 80
  • Krause DS, Van Ellen RA. Tyrosine kinases as targets for cancer therapy. N Engl J Med 2005; 353:172 - 187
  • Ozvegy-Laczka C, Cserepes J, Elkind NB, Sarkadi B. Tyrosine kinase inhibitor resistance in cancer: role of ANC multidrug transporters. Drug Res Update 2005; 8:15 - 26
  • Haouala A, Widmer N, Duchosai MA, Montemurro M, Buclin, Deconsterd LA. Drug interactions with the tyrosine kinase inhibitors imatinib, dasatinib and nilotinib. Blood 2011; 117:e75 - e87
  • Limongi CL, De Souza W, Rozental S. Protein kinase antagonists inhibit invasion of mammlian cells by Fonsecaea pedrosoi. J Med Microbiol 2003; 52:201 - 209
  • Yazdanyar A, Essrmann M, Larsen B. Genistein effects on growth and cell cycle of Candida albicans. J Biomed Sci 2001; 8:153 - 159
  • Filler SG, Swerdloff JN, Hobbs C, Luckett PM. Penetration and damage of endothelial cells by Candida albicans. Infect Immun 1995; 63:976 - 983
  • Luther K, Rohde M, Sturm K, Kotz A, Heeseman J, Ebel F. Characterisation of the phagocytic uptake of Aspergillus conidia by macrophages. Microb Infect 2008; 10:175 - 184
  • Van Erp NP, Gelderblom H, Guchelaar HJ. Clinical pharmacokinetics of tyrosine kinase inhibitors. Cancer Treat Rev 2009; 35:692 - 706
  • Bartoszewska M, Kiel JAKW. The role of macroautophagy in development of filamentous fungi. Antiox Redox Sig 2011; 14:2271 - 2287
  • Richie DL, Askew DS. Autophagy in the filamentous fungus Aspergillus fumigatus. Methods Enzmol 2008; 451:241 - 250
  • Ma J, Jin R, Dobry CJ, Lawson SK, Kumar A. Overexpression of autophagy-related genes inhibits yeast filamentous growth. Autophagy 2007; 3:604 - 609
  • Palmer G, Kelly M, Sturtevant J. Autophagy in the pathogen Candida albicans. Microbiology 2007; 153:51 - 58
  • Roetzer A, Gratz N, Kovrik P, Schuller C. Autophagy supports Candida glabrata survival during phagocytosis. Cell Microbiol 2010; 12:199 - 216
  • Pollack JK, Harris SD, Marten MR. Autophagy in filamentous fungi. Fungal Genet Biol 2009; 46:1 - 8
  • Kikuma T, Arioka M, Katsuhiko K. Autophagy during conidiation and conidial; germination in filamentous fungi. Autophagy 2007; 3:128 - 129
  • Richie DL, Fuller KK, Fortwendel J, Miley MD, McCarthy JW, Feldmesser M, et al. Unexpected link between metal ion deficiency and autopahgy in Aspergillus fumigatus. Eukaryot Cell 2007; 6:2437 - 2447