Antifungal Drugs - The Current Armamentarium and Development of New Agents

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 20

The Fungal Kingdom

Edited by J. Heitman, B. J. Howlett, P. W. Crous, E. H. Stukenbrock, T. Y. James, and N. A. R. Gow


© 2018 American Society for Microbiology, Washington, DC
doi:10.1128/microbiolspec.FUNK-0002-2016

Antifungal Drugs: The Current


Armamentarium and Development
of New Agents
Nicole Robbins,1 Gerard D. Wright,1 and Leah E. Cowen2 44
INTRODUCTION and sub-Saharan Africa (2). Recently, C. gattii was as-
sociated with outbreaks of cryptococcosis in healthy
Importance of Fungal Pathogens individuals on Vancouver Island (5) and other regions
Disease and death caused by fungal infections have
of the Pacific Northwest (6), making Cryptococcus
transitioned from a rare curiosity to a major global
a public health threat to immunocompromised and im-
health problem. Because the vast majority of fungi ca-
munocompetent individuals. Finally, A. fumigatus is a
pable of causing life-threatening infections target indi-
significant cause of invasive infections in individuals
viduals with impaired immunity, recent decades have
with impaired immune function, including those with
witnessed a stark rise in fungal infections due to medi-
neutropenia, solid organ transplant recipients, and pa-
cal interventions such as chemotherapy for cancer and
tients on immunosuppressive therapies such as high-
immunosuppression for organ transplantation and due
dose corticosteroids (2). It is estimated that more than
to the prevalence of HIV infection (1). Pathogenic fungi
200,000 cases of invasive aspergillosis occur each year,
are the causative agents of billions of infections annually,
with staggering mortality rates of up to 50% with treat-
with ∼1.5 million attributable mortalities (2). The public
ment and 100% if left undiagnosed (2). Thus, these
health burden is comparable to that observed with more
chief opportunistic invaders are a leading cause of hu-
notable infectious diseases such as tuberculosis and ma-
man mortality and impose a dire need for effective anti-
laria, yet the impact of fungal infections on human
fungal therapeutics to combat these infections.
health has been largely overlooked (3).
In this article we aim to describe the current arma-
The predominant fungal pathogens of humans include
mentarium of antifungal agents and the most promising
Candida albicans, Cryptococcus neoformans, and Asper-
emerging therapies in the antifungal development pipe-
gillus fumigatus. C. albicans is a common component
line. We also highlight the innovative strategies being
of the human mucosal microbiota and is responsible
leveraged to identify novel antifungal targets and un-
for millions of superficial infections in immunocompe-
cover new therapeutic strategies to combat fungal in-
tent individuals, such as thrush and oropharyngeal can-
fectious disease.
didiasis. It is also the most prevalent fungal etiological
agent of life-threatening invasive infections, with mor-
tality rates approaching 40% despite treatment (1). Challenges for Treatment of Fungal Infections
The CDC has classified Candida species as a serious Fungi are eukaryotic, like their human hosts, limiting
threat to human health due to the dramatic rise in resis- the number of molecular targets that can be selectively
tance to antifungal drugs (4). Cryptococcosis caused by exploited for drug development without the risk of
C. neoformans and the closely related species Crypto- cross-target toxicity. The current arsenal of antifungal
coccus gattii affects over a million individuals annually, compounds approved for the treatment of invasive infec-
with mortality rates as high as 70% in Latin America tions consists of only three classes of antifungal agents

1
Michael G. DeGroote Institute for Infectious Disease Research and Department of Biochemistry and Biomedical Sciences, McMaster University,
Hamilton, ON L8N 3Z5, Canada; 2Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada.

903
904 FUNGI AND THE HUMAN HOST

directed against a limited number of cellular processes. ment of the most serious fungal infections, in large part
Perhaps of greater concern is that there are few discov- due to its broad spectrum of activity.
ery programs devoted to the development of new anti-
fungal therapeutics because of the expectation of limited Pyrimidines
financial return for pharmaceutical companies (2). Most Pyrimidines such as flucytosine (5-fluorocytosine) were
antifungals target ergosterol, the functional fungal ana- first used to treat fungal infections in the 1960s. How-
logue of cholesterol; the biosynthesis of ergosterol; or ever, the rapid onset of resistance precludes the use of
the biosynthesis of (1,3)-β-D-glucan, a major component flucytosine as a monotherapy, and consequently it is
of the fungal cell wall (Fig. 1). This paucity of fungus- only used in combination with other antifungals such
specific targets is problematic because of the prevalence as amphotericin B. This drug combination remains the
of cross-resistance to all drugs with a common target. “gold standard” for treating Cryptococcus infections
Resistance to all widely deployed antifungals has been and other less common invasive mycoses (12). Flucyto-
documented in both the laboratory and the clinic (7) sine is a synthetic fluorinated analogue of cytosine. Upon
and continues to be a growing problem in the medical entry into the cytosol, flucytosine becomes rapidly de-
community. There is a pressing need to bolster the anti- aminated to generate 5-fluorouracil by fungal specific
fungal pipeline, as highlighted by the Generating Anti- cytosine deaminases (13). 5-Fluorouracil acts as a potent
biotic Incentives Now (GAIN) Act (H.R. 2182), which antimetabolite that causes RNA miscoding and inhibits
promises a major effort to reduce the incidence of drug- DNA synthesis (Fig. 1).
resistant fungi and bacteria in the United States through
antimicrobial discovery. Azoles
The azole antifungals encompass compounds with both
imidazole and triazole moieties. They are the most
CLASSES OF ANTIFUNGAL widely deployed class of antifungal in clinical use be-
DRUGS IN CLINICAL USE cause of their impressive safety profile and availability
in both oral and intravenous formulations. For exam-
Polyenes ple, oral azoles are more broadly accessible in resource-
Since the 1950s, the fungicidal polyenes have served limited settings such that oral fluconazole is widely
as a powerful but highly toxic last line of defense for used to treat cryptococcal meningitis in these regions
combating invasive fungal infections. Polyenes are am- despite treatment outcomes being much better with the
phipathic, consisting of a hydrophobic polyene hydro- i.v.-administered combination of amphotericin B and
carbon chain and a hydrophilic polyhydroxyl chain. flucytosine (14). Furthermore, voriconazole is the drug
For decades the prevailing theory was that these mole- of choice for aspergillosis based on its superiority
cules elicited their toxic effects by intercalating into to amphotericin B in a head-to-head clinical trial (15,
ergosterol-containing membranes to form membrane- 16). The azoles consist of a five-membered nitrogen-
spanning channels that cause leakage of cellular com- containing heterocyclic moiety, which binds to an iron
ponents and, ultimately, cell death (8, 9). However, atom in the heme group located in the active site of the
recent detailed structural and biophysical studies high- drug target lanosterol 14-α demethylase (also referred
lighted that polyenes act more like an “ergosterol- to as cytochrome P45014DM and Erg11) (Fig. 1). Azole
sponge,” forming large extramembranous aggregates binding blocks the demethylation of lanosterol, result-
that extract the essential membrane-lipid ergosterol ing in ergosterol depletion and the accumulation of
from the plasma membrane (Fig. 1) (10). Given that toxic sterol intermediates, which ultimately impedes cel-
polyfunctional lipids such as ergosterol serve as central lular growth and division and induces severe membrane
molecular nodes in cellular physiology and function, re- stress (7, 8). Most of the antifungal imidazoles are for-
sistance to the polyenes is strongly evaded and comes at mulated for topical use, as a consequence of toxicity
a significant fitness cost to the fungus (11). The differ- or bioavailability problems that limit their potential as
ential binding affinity of amphotericin B for ergosterol systemic agents. The only agents licensed for treatment
over cholesterol, the mammalian membrane sterol, ac- of invasive fungal disease are triazoles and include
counts for its selectivity; however, amphotericin B does fluconazole, itraconazole, posaconazole, voriconazole,
affect mammalian cells, and nephrotoxicity is a com- and isavuconazole (8, 17). The azoles are typically fun-
mon side effect of clinical usage that can be mitigated gistatic, with the notable exception of voriconazole,
somewhat by liposomal formulations (8). Despite these which has fungicidal activity against A. fumigatus. The
drawbacks, amphotericin B is still used for the treat- fungistatic nature of azoles imposes strong directional
44. ANTIFUNGAL DRUGS: CURRENT ARMAMENTARIUM AND NEW AGENTS 905

Figure 1 Structures and mechanisms of action of clinically relevant antifungal drugs.


(A) The azoles function by targeting the ergosterol biosynthetic enzyme lanosterol de-
methylase, encoded by ERG11 (C. albicans and C. neoformans) or cyp51A and cyp51B
(A. fumigatus), causing a block in the production of ergosterol and the accumulation of a
toxic sterol produced by Erg3. This toxic sterol exerts a severe membrane stress on the cell.
(B) Polyenes such as amphotericin B primarily exist in the form of large extramembranous
aggregates that extract ergosterol from lipid bilayers. (C) Fungal cell walls are composed
of (1,3)-β-D-glucans covalently linked to (1,6)-β-D-glucans as well as chitin, mannans, and
cell wall proteins. The echinocandins act as noncompetitive inhibitors of (1,3)-β-D-glucan
synthase (encoded by FKS1 in C. albicans, C. neoformans, and A. fumigatus) and thereby
cause a loss of cell wall integrity and severe cell wall stress. (D) Pyrimidines such as
flucytosine become rapidly deaminated in the cytosol to generate 5-fluorouracil (5-FU) by
fungal-specific cytosine deaminases. 5-FU acts as a potent antimetabolite that causes RNA
miscoding and inhibits DNA synthesis. Adapted from reference 185 with permission.
906 FUNGI AND THE HUMAN HOST

selection for the evolution of resistance. Moreover, through charge pairing between the amino group of the
several Candida species such as Candida glabrata and amphotericin B and the phosphate group of distearoyl
Candida krusei are intrinsically less susceptible to this phosphatidylglycerol. Tissue concentrations in patients
class of drugs. receiving L-AmB tend to be highest in the liver and
spleen and much lower in the kidneys and lung. In a
Echinocandins rabbit model of hematogenous C. albicans meningo-
The echinocandins are the only new class of antifungal encephalitis, L-AmB accumulates in the brain at higher
to reach the clinic in decades, with three echinocandins levels than standard amphotericin B, significantly de-
currently available for clinical use: caspofungin, mica- creasing fungal burden in this tissue (21). This supports
fungin, and anidulafungin. These compounds are cyclic the preference for L-AmB in treatment of invasive cen-
hexapeptides that act as noncompetitive inhibitors of tral nervous system infections. Amphotericin B colloi-
(1,3)-β-D-glucan synthase, an enzyme involved in cell dal dispersion, marketed as Amphocil and Amphotec,
wall synthesis (Fig. 1). (1,3)-β-D-Glucan is an essential encompasses two molecules of amphotericin B bound
cell wall polysaccharide that provides structural in- to two molecules of cholesteryl sulfate. Similar to the
tegrity. Inhibition of (1,3)-β-D-glucan synthesis culmi- other liposomal formulations, there is reduced drug ac-
nates in a loss of cell wall integrity and severe cell wall cumulation in the kidneys. However, unlike the other
stress (18). The safety profile of these compounds is liposomal formulations, amphotericin B colloidal dis-
impressive and likely attributable to a fungal-specific persion induces a high degree of inflammatory gene
target that is not conserved in mammals. Despite upregulation (20), which was likely a contributing fac-
their potent activity against Candida and Aspergillus, tor related to the adverse side effects observed in sev-
these compounds are completely ineffective at combat- eral clinical studies (22, 23).
ing Cryptococcus infections (18). Echinocandins have In addition to these lipid polyene formulations,
emerged as an important treatment option for candidi- alternative preparations continue to be developed. A
asis, especially in the context of the increasing preva- cochleate formulation of amphotericin B displayed
lence of azole resistance. efficacy in experimental models of candidiasis and as-
pergillosis (24–26). Cochleates are a novel lipid-based
delivery vehicle consisting of crystalline phospholipid-
DEVELOPMENT OF IMPROVED AGENTS cation structures that form spiral lipid sheets. They
WITHIN EXISTING CLASSES represent a new technology platform for oral delivery
of clinically important drugs that possess poor oral
Polyenes bioavailability. Initial biodistribution studies of coch-
In an attempt to improve the therapeutic index of am- leate amphotericin B administered orally in mice found
photericin B, alternative lipid-associated formulations that cochleates deliver significant levels of ampho-
have been developed and incorporated into clinical tericin B to target organs (24, 26). Despite these suc-
use since the 1990s (19). Amphotericin B lipid complex cesses in animal models, clinical trials conducted by
(ABLC) consists of amphotericin B in a complex with BioDelivery Sciences International appear to have been
two lipids: L-α-dimyristoyl phosphatidylcholine and discontinued.
L-α-dimyristoyl phosphatidylglycerol (19). The large For the treatment of most invasive fungal infections,
molecular size of the compound results in rapid uptake an amphotericin B lipid formulation provides a safer
of the drug by macrophages, which distribute the com- alternative than conventional amphotericin B, especially
pound throughout the mononuclear phagocyte sys- in the context of nephrotoxicity. Unfortunately, despite
tem including target organs such as the liver and spleen their use in the clinic for close to 20 years, it is difficult
(19). Consequently, compared with the conventional to conclude which (if any) of these lipid formulations
formulation, there are lower circulating amphotericin B possesses superior in vivo antifungal activity compared
serum concentrations and reduced accumulation in to amphotericin B or compared to each other due to a
the kidneys. Also of benefit, ABLC fails to stimulate paucity of well-designed, randomized trials and also
myriad proinflammatory signaling molecules in vitro due to the multitude of invasive infections caused by
(20). Liposomal amphotericin B (L-AmB), marketed as evolutionarily diverse fungal pathogens.
AmBisome, is composed of a small unilamellar vesicle Due to the pharmacological importance of ampho-
formulation consisting of hydrogenated soy phosphati- tericin B in treating systemic mycoses, medicinal chemists
dylcholine, cholesterol, and distearoyl phosphatidylglyc- have made extensive efforts to generate amphoteri-
erol. Amphotericin B is tightly bound to the liposome cin B derivatives with more favorable pharmacological
44. ANTIFUNGAL DRUGS: CURRENT ARMAMENTARIUM AND NEW AGENTS 907

properties and reduced toxicity. Modification of the the therapeutic effects of a single dose of albaconazole
mycosamine appendage through a double reductive alkyl- in women with acute Candida vulvovaginitis and found
ation generated a semisynthetic molecule with improved that doses of ≥40 mg had greater therapeutic effects
efficacy against amphotericin B-resistant C. albicans than fluconazole at 150 mg (30). Despite the efficacy in
in vitro and reduced hemotoxicity (27). Improvements treatment of vulvovaginitis, it remains unclear whether
to the therapeutic index of amphotericin have also albaconazole will have utility in treatment of invasive
been made in a C2´deoxyamphotericin B derivative fungal infections.
(28). Often the major limitation of these derivatives
is the limited synthetic access, which precludes future Echinocandins
development. Recently, synthetic chemists reported The echinocandins were first introduced into the clinic
urea derivatives of amphotericin B that are easily acces- in 2001. Since this time there has been incentive to de-
sible from the natural product by using a three-step velop novel analogues in this class, primarily to over-
synthesis reaction (29). These derivatives bind ergos- come the current limitation of echinocandins to daily
terol with far greater selectivity than amphotericin B intravenous dosing regimens. Aminocandin (Fig. 2),
in vitro and maintain potent antifungal activity with also referred to as IP960 or HMR3270, is a semisyn-
reduced toxicity in mouse models of disseminated thetic fermentation product from Aspergillus sydowii.
candidiasis (29). Moreover, urea-containing amphoteri- Its long half-life makes infrequent dosing a feasible op-
cin B derivatives strongly evade resistance (29). The tion, and it has shown promising in vitro and in vivo
promising biological activities coupled with favorable activity against Candida and Aspergillus species (33,
synthetic properties suggest bright prospects for these 34). Similarly, Cidara Therapeutics has developed an
amphotericin B derivatives in combating invasive fun- echinocandin derivative, CD 101 IV, which shows a
gal disease. dramatically extended half-life in animal models of
C. albicans infection (35, 36). Preclinical evaluations
Azoles in rats uncovered no hepatotoxicity at doses up to
Given that azoles are the most widely deployed class of 20 mg/kg for 2 weeks (37). Currently, this compound is
antifungal, there is a long history of developing novel in phase I clinical trials (NCT02551549) and has been
analogues with improved efficacy and safety. Isavucon- designated a qualified infectious disease product with
azole was developed as an orally active, water-soluble fast track status from the FDA.
prodrug that is also amenable to intravenous formula- Enfumafungin is a structurally distinct natural prod-
tions (Fig. 2). The prodrug, termed BAL-8557, becomes uct class that is similar to the echinocandins in tar-
cleaved in a reaction catalyzed by plasma esterases in geting glucan synthase (38). The current development
mammalian cells (30). It exhibits a long elimination candidate MK-3118 is an orally active, semisynthetic
half-life, has broad-spectrum activity against Candida derivative of enfumafungin with potent in vitro activity
and Aspergillus species, and has demonstrated good ef- against glucan synthase and potent in vivo activity
ficacy in several clinical trials (31). One such study sup- against Candida and Aspergillus species (Fig. 2) (39,
ported the safety and tolerability of isavuconazole as 40). Thus, novel echinocandin derivatives and distinct
a prophylactic treatment in immunosuppressed patients antifungals targeting glucan synthase activity represent
at high risk of fungal infections (32). This compound promising new treatment options to combat Candida
has undergone phase III clinical trials to evaluate effi- and Aspergillus infections.
cacy in the treatment of Aspergillus infections in renally
impaired patients and those infected with other rare
fungi (clinicaltrials.gov NCT00634049), as well as to WHAT CHEMICAL MATTER SHOULD WE BE
compare efficacy to a caspofungin-voriconazole cother- THINKING ABOUT FOR NEW DRUGS?
apy against Candida infections (NCT00444366 and The optimal chemical space for antifungal compounds
NCT00413218). In 2015, isavuconazole was approved is poorly defined. Unlike antibacterial drugs that are
by the U.S. Food and Drug Administration (FDA) for dominated by microbial natural products, antifungals
the treatment of invasive Aspergillosis and invasive include both synthetic and natural product compounds
mucormycosis. in equal proportion (Fig. 1). In fact, synthetic com-
Albaconazole is another triazole currently under pounds dominate the fungicides used in agriculture.
development with potent broad-spectrum antifungal This should be good news for antifungal drug discovery
activity, good pharmacokinetics, and excellent oral bio- because, in contrast to antibacterial campaigns, exist-
availability (Fig. 2). A phase II clinical study evaluated ing libraries of synthetic compounds in pharmaceutical
908 FUNGI AND THE HUMAN HOST

Figure 2 Structures of compounds with antifungal activity. Chemical structures of anti-


fungal molecules highlighted throughout the review. The description in brackets describes
the molecular target of the chemical compound.
44. ANTIFUNGAL DRUGS: CURRENT ARMAMENTARIUM AND NEW AGENTS 909

companies are likely to be good starting points for NEW ANTIFUNGAL DRUG
antifungal screens. There are challenges in using these TARGETS IN DEVELOPMENT
compounds, however. For example, there are fungal- Our current arsenal of antifungal drugs target either
specific PAIN (pan assay interference) compounds such ergosterol synthesis (azoles), ergosterol itself (polyenes),
as the rhodanines that are promiscuous in antifungal or cell wall synthesis (echinocandins). These targets ac-
campaigns but have a poor likelihood of generating count for a minor fraction of the potential drug targets
good leads in drug development (41). Additionally, the encoded by genomes of fungal pathogens (47). Here we
synthetic compounds that dominate pharmaceutical highlight some of the most promising new antifungal
company libraries are enriched in molecules with phys- drug targets currently being explored for development
icochemico characteristics such as the Lipinski rule of of novel therapeutics.
five (42), which emphasize human targeted drugs. As a
result, compounds identified in antifungal screens from Fungal Sphingolipids
such libraries often overlap with compounds that have Sphingolipids are ubiquitous eukaryotic membrane
bioactivity against human cellular targets. This reflects components that play a central role in signal transduc-
the relatively narrow unique target space between fungi tion, cell regulation, and virulence in pathogenic fungi
and host biology (human, animal, or plant). Threading (48). There are a variety of fungal-specific sphingolipids
the needle between uniquely antifungal targets and the present in the cell membrane and cell wall including
host is significantly challenging, especially if screened glucosylceramides (GlcCer), which are glycosphingo-
compounds are biased toward human biology. Indeed, lipids composed of a glucose unit bound via a glycosyl
as noted below, many highly useful drugs for human linkage to a ceramide moiety. As proof-of-principle,
diseases, such as rapamycin, were first identified in anti- genetic studies have established that targeting sphin-
fungal screens. Not surprisingly then, most antifungal golipid biosynthesis holds promise for blocking fun-
drugs have significant off-target effects that are reflected gal virulence. In C. neoformans, strains lacking GlcCer
in host toxicity. synthase 1 are avirulent in a mouse inhalation infec-
Expanding screens of synthetic molecules to libraries tion model (49). Similarly, deletion of genes required
that typically have not yielded bioactivity in human for sphingolipid biosynthesis in C. albicans renders the
targets (so-called chemical dark matter) can also offer pathogen avirulent (50). These genetic studies predict
an entry point into a selective antifungal target space. that small molecules targeting sphingolipid biosynthesis
A group at Novartis specifically mined such chemical and/or function may serve as novel antifungal agents.
dark matter in a screen for antifungal compounds that High-throughput screening efforts have been fruit-
identified a new compound and a new target, heme ful in identifying small molecules that target sphingo-
biosynthesis (43). lipid biosynthesis. A screen of 49,120 small molecules
Another option is a return to screens of natural to identify inhibitors of fungal sphingolipid GlcCer
products to identify new leads in antifungal drug dis- synthesis identified two compounds: [N´-(3-bromo-4-
covery and development. Screens of microbial natural hydroxybenzylidene)-2-methylbenzohydrazide (BHBM)
product extracts often return polyenes such as ampho- (Fig. 2) and 3-bromo-N´-(3-bromo- 4-hydroxybenzyl-
tericin, but these can often be quickly dereplicated idene) benzohydrazide (D0)] (51). BHBM and D0
based on their characteristic absorption spectra. The were efficacious against C. neoformans in vitro, in an
Merck group has been especially active in this area over intranasal infection model, and in an invasive infection
the past decade, developing new screening technologies model of the central nervous system (51). Paradoxically,
(44) and reporting a series of novel natural products although BHBM and D0 did not show potent anti-
with antifungal activity that modulate new targets fungal activity against C. albicans in vitro, they were
such as poly(A) polymerase (45) and the fungal protea- efficacious in a model of invasive candidiasis (51). Ge-
some (46). nome sequencing of drug-resistant mutants identified
The narrow target space for antifungal drugs, the four candidate targets of BHBM: Apl5, Cos111, Mkk1,
diversity of fungal physiology, and the absence of sys- and Ste2, which are involved in vesicular transport and
tematic physicochemico rules for nontoxic antifungal cell cycle progression (51). Thus, this study has identi-
agents complicate antifungal drug discovery. Neverthe- fied promising compounds that inhibit fungal GlcCer.
less, clever screens for new targets and judicious mining Beyond small molecules, additional strategies to tar-
of natural products and synthetic molecule libraries con- get sphingolipid biosynthesis show considerable prom-
tinue to yield new antifungal leads and offer encourage- ise. Monoclonal antibodies specific to C. neoformans
ment for future success. GlcCer provide protection against C. neoformans infec-
910 FUNGI AND THE HUMAN HOST

tion, likely due to downregulation of the inflammatory inhibitor of Gwt1, a critical acyltransferase required for
response (52). Moreover, treatment with Cerezyme, a the biosynthesis of fungal GPI anchors (62). Sublethal
recombinant enzyme that metabolizes GlcCer, hydro- concentrations of gepinacin or genetic disruption of
lyzes fungal GlcCer in growing C. neoformans cultures, GPI-anchored proteins exposes elevated levels of (1,3)-
disrupts membrane integrity, and exhibits in vivo effi- β-D-glucan on the cell surface, enhancing the host im-
cacy in an inhalation murine model of cryptococcosis mune response against C. albicans (62, 63).
(53). Notably, sphingolipid biosynthesis is also impor-
tant for the host. Compromised sphingolipid biosyn- Farnesyltransferase
thesis in mice confers hypersensitivity to C. albicans Farnesyltranferase is a ubiquitous eukaryotic enzyme
infection due to the inability of immune cells to phago- that catalyzes posttranslational lipidation on the C-
cytose yeast and clear the infection (54). These studies terminus of more than 120 important signaling pro-
reinforce the potential of sphingolipids as an antifungal teins in mammalian cells (64). This acts as a signal,
drug target and highlight the importance of specifically which culminates in the translocation of proteins from
targeting fungal, and not mammalian, sphingolipids. the ER to the plasma membrane. Protein substrates of
farnesyltransferase bear a C-terminal CAAX sequence
GPI Anchor Biosynthesis motif: cysteine (C), two aliphatic residues (AA), and a
Many cell surface proteins are modified at their variable (X) residue. Ras proteins are key modulators
C-terminus by the addition of a glycosylphosphatidyl- of diverse signaling cascades in eukaryotes and are an
inositol (GPI) anchor, which consists of a lipid group, example of proteins modified by farnesyltransferases.
myoinositol, glucosamine, several mannose residues, In pathogenic fungi, Ras signaling pathways regulate
and a phosphoethanolamine group that connects the virulence, morphogenesis, and cell cycle control (65,
GPI anchor to the protein through an amide bond (55). 66). In the model yeast Saccharomyces cerevisiae, dele-
This posttranslational modification results in proteins tion of the gene RAM1 encoding farnesyltransferase
being transported from the endoplasmic reticulum (ER; results in reduced fitness, but it is not essential for via-
the site of modification) to the plasma membrane and bility (67). However, farnesyltransferases are essential
cell wall, where they are directly or indirectly involved for growth and virulence in C. neoformans, C. albi-
in cell wall organization. GPI anchoring is a conserved cans, and A. fumigatus (68–70). Farnesyltransferase
posttranslational modification in eukaryotes, although inhibitors such as manumycin A (Fig. 2), originally de-
the number of mannose groups and the position of side veloped as an anticancer therapeutic, have antifungal
chains on the GPI anchors vary widely between species. activity against C. neoformans and block localization
In C. albicans, deletion of GPI7, a gene with important of C. neoformans Ras1 to the cell membrane (71). Man-
roles in GPI anchor synthesis, results in abnormal mor- umycin A also displays some activity against A. fumi-
phogenesis and reduced virulence (56). In A. fumigatus, gatus and C. albicans (72). Additional farnesyltransferase
transcriptional repression of genes involved in GPI- inhibitors, such as ethylenediamine inhibitor #2 and
anchor biosynthesis results in cell wall defects, abnor- tipifarnib, also show some antifungal activity against
mal morphology, attenuated virulence, and increased C. neoformans, particularly in a mutant deficient in
cell death through autophagy and necrosis (57, 58). capsule production (71). Finally, farnesyltransferase in-
Further, the GPI-anchored protein Ecm33 has important hibitor III blocks the C. albicans morphological transi-
roles in A. fumigatus morphogenesis, cell wall stress re- tion from yeast to hyphae, a virulence trait regulated by
sponse, and virulence (59). Ras signaling (73).
Multiple small molecules that inhibit GPI-anchor Key physical interactions between farnesyltransfer-
biosynthesis have been discovered. The first was iden- ases and their inhibitors have been illuminated through
tified by a screen for molecules that interfere with the structural studies comparing fungal farnesyltransferases
cell wall localization of a GPI-anchored reporter pro- with their human homologs. Using X-ray crystallogra-
tein (60). The initial hit was 1-[4-butylbenzyl] isoquin- phy, conformational differences between these enzymes
olone, a molecule shown to target the acyl transferase from humans and C. neoformans have been identified,
Gwt1 (60). Medicinal chemical-based optimization led supporting feasibility of the development of fungal-
to development of the pyridine-2-amine-based molecule selective farnesyltransferase inhibitors (71). Similarly,
E1210, a compound with potent antifungal activity structural studies of farnesyltransferase enzymes from
against numerous pathogenic fungi (61). Subsequently, A. fumigatus confirmed structural differences from the
a small molecule originally identified as an inducer of human enzyme that are sufficient for species-specific
ER stress, gepinacin, was determined to be a specific targeting (74). Although much of the work with
44. ANTIFUNGAL DRUGS: CURRENT ARMAMENTARIUM AND NEW AGENTS 911

farnesyltransferase inhibitors has focused on their im- Therapeutic exploitation of calcineurin as a tar-
pacts on Ras localization and signaling, mapping the get for antifungal drug development requires fungal-
fungal farnesylome may also uncover novel drug tar- selective agents. Nonimmunosuppressive cyclosporin A
gets for future antifungal development. One example and tacrolimus derivatives retain antifungal activity
is the Ras-related protein RhbA in A. fumigatus, which against C. neoformans, including azole-resistant strains
is important for fungal pathogenesis and vegetative (91). These nonimmunosuppressive analogues also re-
growth in vitro and for virulence in an in vivo model of tain synergistic activity with azoles against C. albicans
infection (75). (84). Chromofungin, a synthetic vasostatin-I-related
peptide, acts as a calcineurin inhibitor and also dis-
Calcineurin plays in vitro activity against A. fumigatus (92). Devel-
A principal regulator of cellular stress responses in opment of effective antifungal therapeutic agents that
all eukaryotes is the Ca2+-calmodulin-activated protein target calcineurin signaling will require crystal struc-
phosphatase calcineurin (76, 77). In pathogenic fungi, tures of calcineurin from fungal pathogens coupled
calcineurin regulates myriad physiological processes in- with structure-guided drug design to enhance potency
cluding cell cycle progression, cation homeostasis, mor- and fungal-selectivity of calcineurin inhibitors or devel-
phogenesis, virulence, and antifungal drug responses opment of agents targeting fungal-specific calcineurin
(76, 77). Calcineurin is required for survival at physio- effectors (77).
logical temperatures in C. neoformans but is dispens-
able for growth in C. albicans and A. fumigatus (77). Hsp90
Hence, the calcineurin signaling pathway has a conserved Hsp90 is an essential molecular chaperone that regu-
role in fungal biology and virulence yet also possesses lates the form and function of diverse client proteins
unique specializations across the fungal genera. (93, 94). As a consequence of its role in stabilizing key
Pharmacological inhibitors of calcineurin function, signal transducers, Hsp90 modulates the relationship
including the natural products cyclosporin A and tac- between genotype and phenotype by acting as both a
rolimus, have potential therapeutic indications. For capacitor and a potentiator for the storage and release
example, these agents are deployed in the clinic as im- of genetic variation (95–97). In pathogenic fungi, tar-
munosuppressants to prevent graft rejection in trans- geting Hsp90 function as an antifungal strategy holds
plant recipients (77). Their immunosuppressive activity considerable promise given that it orchestrates crucial
is due to inhibition of calcineurin-dependent activation cellular responses to drug-induced stress (98–100). In
of a key transcriptional regulator termed the nuclear fac- S. cerevisiae and C. albicans, genetic depletion or phar-
tor of activated T cells (NFAT), which is involved in macological inhibition of Hsp90 impairs the evolution
T cell activation. In fungi, calcineurin inhibitors have of azole resistance and abrogates resistance that has
diverse effects on cellular stress responses and virulence. been acquired through diverse molecular mechanisms
Pharmacological inhibition of calcineurin in C. neofor- (82, 83). Compromise of Hsp90 function also trans-
mans abrogates growth at 37˚C (78), potentiates the forms azoles from fungistatic to fungicidal (82). Phar-
activity of both azoles and echinocandins in C. albicans macological inhibition of Hsp90 in C. neoformans and
(79–83), and renders the fungistatic azoles fungicidal C. gattii inhibits growth and potentiates azole activity
against multiple Candida species (84). Further, cyclo- in vitro (101). Moreover, in C. albicans, C. glabrata,
sporin A and tacrolimus act synergistically with azoles and A. fumigatus, Hsp90 enables basal tolerance and
against C. albicans biofilms both in vitro and in vivo, resistance to the echinocandins. Thus, Hsp90 has a pro-
implicating calcineurin as a key modulator of azole found impact on resistance to multiple antifungal clas-
resistance during biofilm growth (85). In A. fumigatus, ses in diverse fungal pathogens (81, 102–104).
calcineurin inhibitors enhance echinocandin activity Hsp90 regulates responses to antifungal-induced
and transform the fungistatic activity of caspofungin to stress through its downstream effectors calcineurin and
fungicidal (86, 87). Calcineurin inhibitors also show po- protein kinase C signaling (81, 105). In a series of clini-
tent activity against azole- and echinocandin-resistant cal isolates recovered over time from an HIV-infected
strains of A. fumigatus (88). Recently, it was shown patient treated with fluconazole, the basal fluconazole
that calcineurin orchestrates dimorphic transitions, resistance phenotypes of isolates recovered early during
antifungal drug responses, and virulence of the fungal treatment were dependent on Hsp90, calcineurin, and
pathogen Mucor circinelloides (89, 90). Thus, targeting Pkc1 function (82, 105). This dependence gradually
calcineurin provides a logical strategy for combating evolved toward independence of Hsp90, calcineurin,
fungal disease caused by evolutionarily diverse fungi. and Pkc1, corresponding with upregulation of multi-
912 FUNGI AND THE HUMAN HOST

drug transporters (82, 105, 106). Biochemical analysis in vivo rat catheter model of infection, 17-AAG trans-
confirmed that Hsp90 stabilizes the catalytic sub- forms fluconazole from ineffective to highly efficacious
unit of calcineurin and the terminal MAP kinase of without host toxicity in the context of this localized
the Pkc1 cell wall integrity pathway, Mkc1 (81, 105). infection and drug delivery (116). In the context of sys-
Hsp90 is one of the most highly connected hubs in temic fungal infections, toxicity associated with inhibi-
cellular networks, interacting with an estimated 10% tion of host Hsp90 precludes the use of current Hsp90
of the yeast proteome (107, 108). Chemical genomic inhibitors as antifungals and demands the development
studies in C. albicans identified a multitude of Hsp90 of fungal-selective Hsp90 inhibitors or agents that tar-
genetic interactors important for cellular responses to get fungal-specific regulators of Hsp90 function.
azoles and echinocandins (109), suggesting that Hsp90
may have pleiotropic effects on circuitry governing Acetyltransferases and Deacetylases
drug resistance. Lysine deacetylases (KDACs) and lysine acetyltrans-
Hsp90 is also a powerful therapeutic target given ferases (KATs) catalyze the removal or addition of ace-
its function in orchestrating central virulence traits tyl groups from the ε-amino group of lysines. Such
such as fungal morphogenesis. In C. albicans the ability modifications in core histones result in changes in chro-
to transition between distinct morphological states is matin structure and gene expression (117, 118). Con-
correlated with virulence (50) and is tightly regulated sequently, KDACs and KATs have pleiotropic effects
by diverse environmental cues coupled with elevated on cellular signaling and stress responses in fungi. The
temperatures of at least 37˚C (7). Genetic or pharmaco- broad-spectrum KDAC inhibitor trichostatin A poten-
logical inhibition of Hsp90 function induces filamenta- tiates azole activity in C. albicans (119, 120). Further,
tion by relieving its repression on Ras1-protein kinase C. albicans clinical isolates are sensitive to nicotin-
A signaling (110, 111). Elevated temperatures similarly amide, an inhibitor of NAD+-dependent KDAC com-
relieve Hsp90-mediated repression of filamentation as plexes, and nicotinamide reduces fungal kidney burden
the chaperone becomes overwhelmed with global pro- in a mouse model of disseminated candidiasis (121).
tein misfolding in the cell (110, 112). To add additional Nicotinamide is also active against other Candida spe-
complexity, Hsp90 also regulates C. albicans morpho- cies and against A. fumigatus and Aspergillus nidulans,
genesis through the cyclin Pcl1, the cyclin-dependent suggesting broad antifungal properties (121). Fur-
kinases Pho85 and Cdc28, and the transcription fac- ther, a molecule thought to inhibit the KDAC Hos2,
tor Hms1 (113, 114). Intriguingly, genetic depletion of MGCD290 (Mirati Therapeutics, San Diego, CA), dis-
Hsp90 in A. fumigatus has the opposite effect in that it plays synergistic activity with azoles and echinocandins
decreases hyphal growth and leads to severe defects in against diverse C. albicans drug-resistant clinical isolates
germination and conidiation (102). Given its intimate (122, 123). These studies were restricted to in vitro anal-
and complex role in governing fungal morphogenesis in ysis, but in vivo studies coupled with preliminary clini-
multiple species, targeting Hsp90 holds promise as an cal trials have supported the use of MGCD290 in
antivirulence strategy to combat fungal infection. combination with fluconazole to treat C. albicans infec-
The impact of Hsp90 inhibition on fungal virulence tions (124). Thus, these findings suggest considerable
and antifungal efficacy has been explored using a vari- promise for the use of KDAC inhibitors in combination
ety of in vivo infection models. In Galleria mellonella with current antifungals in combating invasive fungal
models of fungal pathogenesis, combination therapy disease.
with Hsp90 inhibitors that have been in clinical devel- In addition to their roles in modifying histones,
opment for cancer, such as 17-AAG (Fig. 2), with either KDACs also deacetylate a spectrum of nonhistone pro-
caspofungin or fluconazole improves survival upon in- teins, such as Hsp90. Acetylation of Hsp90 provides a
fection with A. fumigatus or C. albicans, respectively mechanism of posttranslational control that governs
(115). Furthermore, in murine models of C. albicans dis- the emergence and maintenance of Hsp90-dependent
seminated disease, genetic compromise of fungal HSP90 azole resistance (120). In S. cerevisiae, Hsp90 is acety-
expression reduces virulence (110) and enhances the lated on lysine 27 and 270, residues that are regulated
therapeutic efficacy of fluconazole and caspofungin by KDACs Rpd3 and Hda1 (120). Genetic or pharma-
(81, 115). Similarly, genetic compromise of A. fumi- cological inhibition of both Hda1 and Rpd3 blocks the
gatus Hsp90 abrogates virulence in a murine model of physical interaction between Hsp90 and calcineurin,
invasive aspergillosis (104). In a Caenorhabditis elegans thereby impeding calcineurin function (120). Deletion
model of cryptococcal infection, Hsp90 inhibition en- of both HDA1 and RPD3 does not increase global
hances the activity of fluconazole (101). Finally, in an Hsp90 acetylation levels, suggesting that additional
44. ANTIFUNGAL DRUGS: CURRENT ARMAMENTARIUM AND NEW AGENTS 913

KDACs orchestrate Hsp90 deacetylation. One candi- models of candidiasis (Fig. 2) (45). Inhibitors of leucine
date KDAC is Hos2, which interacts genetically with tRNA synthases, such as tavaborole (AN2690), are
Hsp90 under diverse environmental conditions in C. albi- highly selective against Trichophyton species (137, 138)
cans (109). In A. fumigatus, Hsp90 lysine residues im- and are in clinical development to treat onychomycosis
portant for drug resistance and virulence are conserved (14). Finally, inhibitors targeting the 26S proteasome
with those identified in S. cerevisiae (120, 125). Thus, (fellutamides, Fig. 2), translation elongation (yefa-
by influencing gene expression through histone modifi- fungin), cyclic AMP homeostasis (campafungin), and
cation and controlling the function of key signal trans- microtubule dynamics (12-deoxy-hamigerone) are all
ducers, KDAC inhibitors hold promise as antifungal unexploited areas that may represent the next break-
therapeutics. through in novel antifungal scaffold (14, 44, 46). While
KATs also modulate azole resistance. Genetic impair- it is impossible to predict which, if any, of the targets
ment of C. albicans ADA2, which encodes a component highlighted above will prove to be the next clinically
of the Spt-Ada-Gcn5-acetyltransferase (SAGA) coactiva- exploited antifungal target, the portfolio of distinct
tor complex, confers hypersensitivity to fluconazole due prospects provides renewed hope for the development
to impaired upregulation of the efflux pumps Cdr1 and of novel antifungal treatments.
Mdr1 (126). Further, deletion of the C. albicans KAT
gene RTT109 confers hypersusceptibility to macrophages,
altered metabolic gene expression, and induction of a COMBINATION THERAPY TO EXPAND
weaker inflammatory response, culminating in attenuated THE REPERTOIRE OF DRUG TARGETS
virulence in a murine infection model (121, 127). Deletion Although advances are being made to improve our
of RTT109 also confers increased sensitivity to echino- current antifungal arsenal and to identify novel drug
candins and to other genotoxic stresses such as hydroxy- targets, the rate at which antimicrobials are being pro-
urea and methyl methane sulfonate (121). duced is vastly outpaced by the rate at which resis-
tance is emerging (139, 140). A promising strategy for
Other Antifungal Targets combating drug-resistant fungi is through combination
The TOR protein kinases are global regulators of cellu- therapy. Drug combination therapy has benefits such as
lar growth in response to nutrient cues, including amino enhanced efficacy and specificity when compared to in-
acids (128). In S. cerevisiae, inhibition of TOR signal- dividual drug treatments and can impede the evolution
ing has a multitude of effects including induction of of resistance (141, 142). Further, by carefully selecting
autophagy, inhibition of translation, and repression specific drug combinations, microbial drug resistance
of ribosomal gene expression, as well as upregulation may be not only neutralized but also reversed through
of genes involved in the retrograde response, nitrogen a process called selection inversion (143). Combina-
catabolite response, and stress response (128–130). tion therapy provides the foundation for treatment of
TOR can be inhibited pharmacologically by the immu- other infectious diseases such as HIV (144), tuberculo-
nosuppressant rapamycin (sirolimus). Rapamycin is a sis (145), and malaria (146). Despite the therapeutic
natural product of the bacterium Streptomyces hygro- potential, there is only one combination that has been
scopicus, originally identified in a screen for antimi- well validated in clinical trials for fungal infections: the
crobial activity against C. albicans and later found to combination of amphotericin B and flucytocine as the
have potent immunosuppressive activity (131). Pioneer- gold standard to treat cryptococcosis (12). Combina-
ing genetic studies in S. cerevisiae identified FKBP12, tion therapy also has the potential to unveil myriad
Tor1, and Tor2 as the direct targets of rapamycin additional antifungal targets given that fungal biology
(132). Rapamycin inhibits growth of diverse fungi, in- is controlled by a highly interconnected and function-
cluding C. neoformans, C. albicans, A. fumigatus, Fusa- ally redundant network of biological interactions (147,
rium oxysporum, and Penicillium spp. (133, 134) and 148). The yeast deletion project revealed that only
abrogates erg3-mediated azole resistance (135). Ana- ∼1,000 of the ∼6,000 genes in the S. cerevisiae genome
logues of rapamycin with reduced immunosuppressive are essential for viability (149). However, a tour de
activity retain antifungal activity in vitro (136). force systematic analysis of ∼5.4 million pairs of gene
There are several other antifungal agents in develop- deletions identified ∼170,000 negative genetic interac-
ment that target a variety of other cellular processes. tions, including 10,000 synthetic lethal interactions
Parnafungins inhibit poly(A) polymerase and display po- (148). Synthetic lethality represents an extreme nega-
tent broad-spectrum activity against clinically relevant tive genetic interaction in which two mutations, causing
Candida species and therapeutic efficacy in murine little to no fitness defect individually, result in a lethal
914 FUNGI AND THE HUMAN HOST

double mutant phenotype (150). The potential for enzymes (140). A recent study with C. glabrata iden-
harnessing synthetic lethality provides a new avenue for tified a small molecule (iKIX1) (Fig. 2) that inhibits
antimicrobial drug discovery through the use of combi- azole efflux pump expression by disrupting the interac-
nation agents. tion between the mediator complex and the transcrip-
There is precedent for leveraging the power of ge- tional activator Pdr1 (159). Follow-up in vivo studies
netic interactions to predict combination drug therapy. supported the use of iKIX1 to potentiate fluconazole ef-
In S. cerevisiae, FKS1 and its paralog FKS2 encode the ficacy against fluconazole-resistant C. glabrata in both
biosynthetic enzyme for (1,3)-β-D-glucan synthesis and metazoan and mammalian models of infection (159).
the molecular target of the echinocandins. FKS1 and A complementary approach to identify synergistic
FKS2 are synthetically lethal, in keeping with echino- drug interactions employed in both academia and in-
candin efficacy. FKS1 is also synthetically lethal with dustry utilizes high-throughput screening of large com-
CHS3, a chitin synthase required for the synthesis of pound collections. Molecules that enhance the activity
the cell wall component chitin (148), and inhibitors of of established antifungals against S. cerevisiae have
chitin synthases, such as nikkomycin, are synergistic been identified by chemical biology screens (160, 161)
with caspofungin against numerous fungal pathogens and computational methods (162–164). Recent large-
(151, 152). FKS1 is also synthetically lethal with CNB1, scale screening efforts have extended the strategy of
the regulatory subunit with calcineurin, and as high- chemical synthetic lethality to the fungal pathogens
lighted above, calcineurin inhibitors exert potent syn- C. albicans and C. neoformans. Two such studies ex-
ergy with echinocandins against diverse fungi (77). amined the potential of drugs approved for other ther-
While these examples support the fact that genetic in- apeutic indications to potentiate current antifungal
teractions underpin the network response to compound agents (157, 165). Compounds that increase azole effi-
combinations, the complexities of chemical action and cacy were found to often target membrane function or
genetic network density seem to preclude the predic- sphingolipid biosynthesis (165). One such example is
tion of synergism on a genome-wide scale (153). Thus, the antimycobacterium compound clofazimine, which
additional factors must be employed to identify effec- induces cell membrane stress in fungi (157). A combi-
tive antifungal combinations. nation of clofazimine and caspofungin was efficacious
In addition to leveraging genetic interaction land- in vivo against C. albicans in a G. mellonella model
scapes to predict chemical synergy, target-specific in- of infection (157). These studies illustrate the potential
hibitors may be screened ad hoc for synergistic activity of combining current antifungal treatments with repur-
against specific antifungal agents. We explored many posed medications.
of these examples above, as with the capacity of inhibi-
tors of Hsp90 (98, 99), calcineurin (77), KDACs (120,
123), and TOR (136), to potentiate antifungals against CHEMICAL GENOMICS TO LINK
fungal pathogens. Other notable examples include po- BIOACTIVES TO CELLULAR TARGETS
tentiation of azole activity by pharmacological inhibi- A variety of genetic and genomic approaches have been
tion of ADP-ribosylation factors (154), protein kinase developed to uncover the molecular target for a com-
C (105), and protein translation (155). Further, tamoxi- pound whose mechanism of action remains elusive. Tra-
fen and other triphenylethylene-based estrogen recep- ditionally, forward chemical-genetic approaches have
tors have antifungal activity against C. neoformans been used to identify targets by selection of resistant
(156) and potentiate the activity of azoles, polyenes, mutants coupled with plasmid-based library comple-
and echinocandins in diverse fungal species (157). mentation, as well as gene and whole-genome sequenc-
Tamoxifen inhibits the C. neoformans calmodulin pro- ing (166).
tein Cam1, which activates calcineurin (158). Given that More recently, reverse chemical-genetic approaches
Cam1 is fungal-specific, optimization of the triphenyl- have become widely used to identify the mechanism
ethylene scaffold provides a promising path for anti- of action of bioactive agents. In S. cerevisiae, haplo-
fungal drug development. insufficiency profiling screens were developed to iden-
Small molecules that target resistance determinants tify targets of bioactive compounds de novo based on
can be effectively paired with currently available anti- the principle that reducing the dosage of a drug tar-
microbials to combat drug-resistant isolates. Examples get confers hypersensitivity to the drug. In these assays,
of such drug combinations in the clinic include drugs a genome-scale collection of heterozygous deletion
such as amoxicillin, a β-lactam antibiotic, with clavu- strains are pooled, grown in the presence of compound,
lanic acid, an inhibitor of β-lactamase drug-resistant and sampled over time (167, 168). Each heterozygous
44. ANTIFUNGAL DRUGS: CURRENT ARMAMENTARIUM AND NEW AGENTS 915

strain has a unique barcode sequence that enables de- complexity random mutagenesis (or variomic) libraries
convolution of strain identity in pooled screens (169). covering ∼90% of the S. cerevisiae genome. Screening
Quantitative analysis of strain fitness can be achieved by this library against test compounds allows for systematic
next-generation sequencing or microarray technology to discovery of resistance-conferring mutations, thereby de-
monitor barcode abundance (167, 168). Homozygous fining residues critical for drug binding and identifying
deletion profiling (HOP) provides a complementary as- potential mechanisms of resistance (177). Research com-
say to investigate the mode of action of compounds that bining chemogenomic and variomic tools has led to the
lack an essential protein target (for example, DNA or identification of two novel antifungal geranylgeranyl-
lipids) or for those compounds with redundant protein transferase I inhibitors, highlighting the importance of an
targets (166). Further, HOP reveals the genes important integrated platform for drug target identification (178).
for buffering the cellular response to a drug. For exam-
ple, compounds that target DNA generate HOP profiles
that identify nonessential DNA-repair genes and com- OUTLOOK
plexes required for surviving DNA damage (170). It is clear that creative and focused efforts are urgently
Large chemogenomic screening efforts have unveiled needed to bolster the antifungal pipeline and improve
chemical-genetic interaction maps for thousands of clinical outcome for the millions of people with life-
small molecules and revealed how eukaryotic cells threatening fungal infections worldwide. There are new
respond to cellular perturbations. An intensive study strategies on the horizon to expand the development
with 1,144 haploinsufficiency profiling and 418 HOP opportunities beyond targeting essential genes or cellu-
experiments identified growth phenotypes for mutants lar functions. A complementary approach is to target
covering 97% of the S. cerevisiae genome (171). This proteins that are required for pathogen growth in the
expands our knowledge of yeast physiology and sug- host or for virulence. Identifying nonessential virulence
gests that most genes are conditionally essential with factors opens new opportunities for chemical diversity
functions that can be probed by small molecules. Che- in therapeutic drugs, because the cognate inhibitors
mogenomic profiles generated for 3,250 compounds are not typically explored by conventional screening
in S. cerevisiae revealed that the cellular response to approaches. Although the potential utility of targeting
small molecules is largely restricted to a network of nonessential genes has only recently been appreciated,
45 chemogenomic signatures (172). These signatures there are already examples of antibiotics that target es-
highlight the fundamental small molecule response sys- sential structures rather than essential proteins, as with
tem in eukaryotic cells and provide a resource for daptomycin and the Gram-positive cell wall (179). Fur-
the exploration of relationships between genes, biolo- ther, the diphtheria and tetanus vaccines, as well as the
gical processes, and chemical structures (172). Using antitoxin for botulinum toxin, are examples of life-
chemical-genomic datasets, researchers recently devel- saving treatments targeting nonessential virulence fac-
oped a machine-learning-based prediction algorithm tors (180, 181). Targeting virulence factors offers many
capable of predicting chemical synergies from large benefits including expanding the repertoire of antifun-
chemical libraries (153). This algorithm achieves a gal targets, minimizing effects on the host mycobiome,
6-fold enrichment in predicting synergistic chemical and reducing selection pressure for the evolution of
pairs with activity against fungi compared with chance drug resistance (182, 183). With advances in genomics
selection alone, providing an effective platform for technologies and structure-guided drug design, as well
identifying antifungal chemical combinations. as having antifungals now granted “orphan status” by
Enabled by the development of powerful functional the FDA and a priority in the GAIN act (H.R. 2182)
genomics resources, analogous tools for chemical biology to facilitate clinical trials (184), there is great potential
are now available for the fungal pathogens C. albicans to foster stronger relationships between academia and
(173, 174) and C. neoformans (175). It is becoming industry to accelerate antifungal drug discovery.
abundantly clear that gene essentiality and chemical-
Citation. Robbins N, Wright GD, Cowen LE. 2016. Antifun-
genetic responses diverge rapidly over evolutionary time gal drugs: the current armamentarium and development of
(175, 176), underscoring the importance of performing new agents. Microbiol Spectrum 4(5):FUNK-0002-2016.
chemical genomic analyses directly in fungal pathogens.
In addition to chemogenomic profiling, the develop- References
ment of a functional variomics tool provides a com- 1. Pfaller MA, Diekema DJ. 2010. Epidemiology of inva-
plementary approach to elucidate drug mode of action sive mycoses in North America. Crit Rev Microbiol 36:
(177). This technology applies plasmid-based, high- 1–53.
916 FUNGI AND THE HUMAN HOST

2. Brown GD, Denning DW, Gow NA, Levitz SM, Netea P, Lortholary O, Sylvester R, Rubin RH, Wingard JR,
MG, White TC. 2012. Hidden killers: human fungal in- Stark P, Durand C, Caillot D, Thiel E, Chandrasekar
fections. Sci Transl Med 4:165rv13. PH, Hodges MR, Schlamm HT, Troke PF, de Pauw B,
3. Armstrong-James D, Meintjes G, Brown GD. 2014. A Invasive Fungal Infections Group of the European
neglected epidemic: fungal infections in HIV/AIDS. Organisation for Research and Treatment of Cancer
Trends Microbiol 22:120–127. and the Global Aspergillus Study Group. 2002. Voricon-
4. CDC. 2013. Antibiotic Resistance Threats in the United azole versus amphotericin B for primary therapy of in-
States, 2013. CDC, Atlanta, GA. vasive aspergillosis. N Engl J Med 347:408–415.
5. Kidd SE, Hagen F, Tscharke RL, Huynh M, Bartlett 17. Miceli MH, Kauffman CA. 2015. Isavuconazole: a new
KH, Fyfe M, Macdougall L, Boekhout T, Kwon-Chung broad-spectrum triazole antifungal agent. Clin Infect
KJ, Meyer W. 2004. A rare genotype of Cryptococcus Dis 61:1558–1565.
gattii caused the cryptococcosis outbreak on Vancouver 18. Denning DW. 2003. Echinocandin antifungal drugs.
Island (British Columbia, Canada). Proc Natl Acad Sci Lancet 362:1142–1151.
USA 101:17258–17263. 19. Hamill RJ. 2013. Amphotericin B formulations: a com-
6. Byrnes EJ III, Li W, Lewit Y, Ma H, Voelz K, Ren P, parative review of efficacy and toxicity. Drugs 73:919–
Carter DA, Chaturvedi V, Bildfell RJ, May RC, Heitman 934.
J. 2010. Emergence and pathogenicity of highly virulent 20. Simitsopoulou M, Roilides E, Dotis J, Dalakiouridou
Cryptococcus gattii genotypes in the northwest United M, Dudkova F, Andreadou E, Walsh TJ. 2005. Differ-
States. PLoS Pathog 6:e1000850. ential expression of cytokines and chemokines in human
7. Shapiro RS, Robbins N, Cowen LE. 2011. Regulatory monocytes induced by lipid formulations of ampho-
circuitry governing fungal development, drug resistance, tericin B. Antimicrob Agents Chemother 49:1397–1403.
and disease. Microbiol Mol Biol Rev 75:213–267. 21. Groll AH, Giri N, Petraitis V, Petraitiene R, Candelario
8. Ostrosky-Zeichner L, Casadevall A, Galgiani JN, Odds M, Bacher JS, Piscitelli SC, Walsh TJ. 2000. Compara-
FC, Rex JH. 2010. An insight into the antifungal pipe- tive efficacy and distribution of lipid formulations of
line: selected new molecules and beyond. Nat Rev Drug amphotericin B in experimental Candida albicans infec-
Discov 9:719–727. tion of the central nervous system. J Infect Dis 182:
9. Gruszecki WI, Gagoś M, Hereć M, Kernen P. 2003. 274–282.
Organization of antibiotic amphotericin B in model 22. Timmers GJ, Zweegman S, Simoons-Smit AM, van
lipid membranes. A mini review. Cell Mol Biol Lett 8: Loenen AC, Touw D, Huijgens PC. 2000. Amphotericin
161–170. B colloidal dispersion (Amphocil) vs fluconazole for the
10. Anderson TM, Clay MC, Cioffi AG, Diaz KA, Hisao prevention of fungal infections in neutropenic patients:
GS, Tuttle MD, Nieuwkoop AJ, Comellas G, Maryum data of a prematurely stopped clinical trial. Bone Mar-
N, Wang S, Uno BE, Wildeman EL, Gonen T, Rienstra row Transplant 25:879–884.
CM, Burke MD. 2014. Amphotericin forms an extra- 23. Herbrecht R, Letscher V, Andres E, Cavalier A. 1999.
membranous and fungicidal sterol sponge. Nat Chem Safety and efficacy of amphotericin B colloidal disper-
Biol 10:400–406. sion. An overview. Chemotherapy 45(Suppl 1):67–76.
11. Vincent BM, Lancaster AK, Scherz-Shouval R, 24. Delmas G, Park S, Chen ZW, Tan F, Kashiwazaki R,
Whitesell L, Lindquist S. 2013. Fitness trade-offs restrict Zarif L, Perlin DS. 2002. Efficacy of orally delivered
the evolution of resistance to amphotericin B. PLoS Biol cochleates containing amphotericin B in a murine model
11:e1001692. of aspergillosis. Antimicrob Agents Chemother 46:2704–
12. Day JN, Chau TT, Wolbers M, Mai PP, Dung NT, Mai 2707.
NH, Phu NH, Nghia HD, Phong ND, Thai CQ, Thai 25. Zarif L, Graybill JR, Perlin D, Najvar L, Bocanegra R,
H, Chuong LV, Sinh DX, Duong VA, Hoang TN, Diep Mannino RJ. 2000. Antifungal activity of amphotericin
PT, Campbell JI, Sieu TP, Baker SG, Chau NV, Hien B cochleates against Candida albicans infection in a
TT, Lalloo DG, Farrar JJ. 2013. Combination antifun- mouse model. Antimicrob Agents Chemother 44:1463–
gal therapy for cryptococcal meningitis. N Engl J Med 1469.
368:1291–1302. 26. Santangelo R, Paderu P, Delmas G, Chen ZW, Mannino
13. Loyse A, Dromer F, Day J, Lortholary O, Harrison TS. R, Zarif L, Perlin DS. 2000. Efficacy of oral cochleate-
2013. Flucytosine and cryptococcosis: time to urgently amphotericin B in a mouse model of systemic candidia-
address the worldwide accessibility of a 50-year-old an- sis. Antimicrob Agents Chemother 44:2356–2360.
tifungal. J Antimicrob Chemother 68:2435–2444. 27. Paquet V, Carreira EM. 2006. Significant improve-
14. Roemer T, Krysan DJ. 2014. Antifungal drug develop- ment of antifungal activity of polyene macrolides by
ment: challenges, unmet clinical needs, and new ap- bisalkylation of the mycosamine. Org Lett 8:1807–
proaches. Cold Spring Harb Perspect Med 4:a019703. 1809.
15. Denning DW, Ribaud P, Milpied N, Caillot D, Herbrecht 28. Wilcock BC, Endo MM, Uno BE, Burke MD. 2013.
R, Thiel E, Haas A, Ruhnke M, Lode H. 2002. Efficacy C2´-OH of amphotericin B plays an important role in
and safety of voriconazole in the treatment of acute in- binding the primary sterol of human cells but not yeast
vasive aspergillosis. Clin Infect Dis 34:563–571. cells. J Am Chem Soc 135:8488–8491.
16. Herbrecht R, Denning DW, Patterson TF, Bennett JE, 29. Davis SA, Vincent BM, Endo MM, Whitesell L,
Greene RE, Oestmann JW, Kern WV, Marr KA, Ribaud Marchillo K, Andes DR, Lindquist S, Burke MD. 2015.
44. ANTIFUNGAL DRUGS: CURRENT ARMAMENTARIUM AND NEW AGENTS 917

Nontoxic antimicrobials that evade drug resistance. Nat by two international methods (CLSI and EUCAST).
Chem Biol 11:481–487. J Antimicrob Chemother 68:858–863.
30. Pasqualotto AC, Denning DW. 2008. New and emerg- 41. Pouliot M, Jeanmart S. 2016. Pan assay interference
ing treatments for fungal infections. J Antimicrob compounds (PAINS) and other promiscuous compounds
Chemother 61(Suppl 1):i19–i30. in antifungal research. J Med Chem 59:497–503.
31. Slavin MA, Thursky KA. 2016. Isavuconazole: a role 42. Lipinski CA. 2004. Lead- and drug-like compounds:
for the newest broad-spectrum triazole. Lancet 387: the rule-of-five revolution. Drug Discov Today Technol
726–728. 1:337–341.
32. Cornely OA, Böhme A, Schmitt-Hoffmann A, Ullmann 43. Wassermann AM, Lounkine E, Hoepfner D, Le Goff G,
AJ. 2015. Safety and pharmacokinetics of isavuconazole King FJ, Studer C, Peltier JM, Grippo ML, Prindle V,
as antifungal prophylaxis in acute myeloid leukemia Tao J, Schuffenhauer A, Wallace IM, Chen S, Krastel P,
patients with neutropenia: results of a phase 2, dose Cobos-Correa A, Parker CN, Davies JW, Glick M. 2015.
escalation study. Antimicrob Agents Chemother 59: Dark chemical matter as a promising starting point for
2078–2085. drug lead discovery. Nat Chem Biol 11:958–966.
33. Warn PA, Sharp A, Morrissey G, Denning DW. 2010. 44. Roemer T, Xu D, Singh SB, Parish CA, Harris G,
Activity of aminocandin (IP960; HMR3270) compared Wang H, Davies JE, Bills GF. 2011. Confronting the
with amphotericin B, itraconazole, caspofungin and challenges of natural product-based antifungal discov-
micafungin in neutropenic murine models of dissemi- ery. Chem Biol 18:148–164.
nated infection caused by itraconazole-susceptible and 45. Jiang B, Xu D, Allocco J, Parish C, Davison J, Veillette
-resistant strains of Aspergillus fumigatus. Int J Anti- K, Sillaots S, Hu W, Rodriguez-Suarez R, Trosok S,
microb Agents 35:146–151. Zhang L, Li Y, Rahkhoodaee F, Ransom T, Martel N,
34. Warn PA, Sharp A, Morrissey G, Denning DW. 2005. Wang H, Gauvin D, Wiltsie J, Wisniewski D, Salowe S,
Activity of aminocandin (IP960) compared with ampho- Kahn JN, Hsu MJ, Giacobbe R, Abruzzo G, Flattery A,
tericin B and fluconazole in a neutropenic murine model Gill C, Youngman P, Wilson K, Bills G, Platas G, Pelaez
of disseminated infection caused by a fluconazole- F, Diez MT, Kauffman S, Becker J, Harris G, Liberator
resistant strain of Candida tropicalis. J Antimicrob P, Roemer T. 2008. PAP inhibitor with in vivo efficacy
Chemother 56:590–593. identified by Candida albicans genetic profiling of natu-
35. James K, Laudeman C, Malkar N, Krishnan R, ral products. Chem Biol 15:363–374.
Polowy K. 2015. Structure-activity relationship of a 46. Xu D, Ondeyka J, Harris GH, Zink D, Kahn JN, Wang
series of echinocandins and the discovery of CD101, a H, Bills G, Platas G, Wang W, Szewczak AA, Liberator
highly stable and soluble, once-weekly novel echino- P, Roemer T, Singh SB. 2011. Isolation, structure, and
candin. Cidara Therapeutics. Abstr ICAAC/ICC, San biological activities of Fellutamides C and D from an
Diego, abstr F-750. undescribed Metulocladosporiella (Chaetothyriales) us-
36. Zhao Y, Kolesnikova I, Dolgov E, Perlin D. 2015. Effi- ing the genome-wide Candida albicans fitness test. J Nat
cacy of CD101 to treat echinocandin-resistant Candida Prod 74:1721–1730.
albicans in a murine model of invasive Candidiasis. 47. Hopkins AL, Groom CR. 2002. The druggable genome.
Cidara Therapeutics. Abstr ICAAC/ICC, San Diego, Nat Rev Drug Discov 1:727–730.
abstr F-748. 48. Thevissen K, Kristensen HH, Thomma BP, Cammue
37. Ong V, Hough G, Schlosser M, Bartizal K, Balkovec J, BP, François IE. 2007. Therapeutic potential of anti-
James K, Krishnan R. 2015. Preclinical evaluation fungal plant and insect defensins. Drug Discov Today
shows CD101, a novel echinocandin, is highly stable 12:966–971.
with no hepatotoxicity in rats. Cidara Therapeutics. 49. Rittershaus PC, Kechichian TB, Allegood JC, Merrill
Abstr ICAAC/ICC, San Diego, abstr A015. AH Jr, Hennig M, Luberto C, Del Poeta M. 2006.
38. Onishi J, Meinz M, Thompson J, Curotto J, Dreikorn S, Glucosylceramide synthase is an essential regulator of
Rosenbach M, Douglas C, Abruzzo G, Flattery A, Kong pathogenicity of Cryptococcus neoformans. J Clin In-
L, Cabello A, Vicente F, Pelaez F, Diez MT, Martin vest 116:1651–1659.
I, Bills G, Giacobbe R, Dombrowski A, Schwartz R, 50. Noble SM, French S, Kohn LA, Chen V, Johnson AD.
Morris S, Harris G, Tsipouras A, Wilson K, Kurtz MB. 2010. Systematic screens of a Candida albicans homo-
2000. Discovery of novel antifungal (1,3)-beta-D-glucan zygous deletion library decouple morphogenetic switch-
synthase inhibitors. Antimicrob Agents Chemother 44: ing and pathogenicity. Nat Genet 42:590–598.
368–377. 51. Mor V, Rella A, Farnoud AM, Singh A, Munshi M, Bryan
39. Pfaller MA, Messer SA, Motyl MR, Jones RN, A, Naseem S, Konopka JB, Ojima I, Bullesbach E,
Castanheira M. 2013. In vitro activity of a new oral Ashbaugh A, Linke MJ, Cushion M, Collins M, Ananthula
glucan synthase inhibitor (MK-3118) tested against HK, Sallans L, Desai PB, Wiederhold NP, Fothergill AW,
Aspergillus spp. by CLSI and EUCAST broth micro- Kirkpatrick WR, Patterson T, Wong LH, Sinha S, Giaever
dilution methods. Antimicrob Agents Chemother 57: G, Nislow C, Flaherty P, Pan X, Cesar GV, de Melo
1065–1068. Tavares P, Frases S, Miranda K, Rodrigues ML, Luberto
40. Pfaller MA, Messer SA, Motyl MR, Jones RN, C, Nimrichter L, Del Poeta M. 2015. Identification of a
Castanheira M. 2013. Activity of MK-3118, a new oral new class of antifungals targeting the synthesis of fungal
glucan synthase inhibitor, tested against Candida spp. sphingolipids. MBio 6:e00647-15.
918 FUNGI AND THE HUMAN HOST

52. Rodrigues ML, Shi L, Barreto-Bergter E, Nimrichter L, 2015. Abolishing cell wall glycosylphosphatidylinositol-
Farias SE, Rodrigues EG, Travassos LR, Nosanchuk JD. anchored proteins in Candida albicans enhances recog-
2007. Monoclonal antibody to fungal glucosylceramide nition by host dectin-1. Infect Immun 83:2694–2704.
protects mice against lethal Cryptococcus neoformans 64. Berndt N, Hamilton AD, Sebti SM. 2011. Targeting
infection. Clin Vaccine Immunol 14:1372–1376. protein prenylation for cancer therapy. Nat Rev Cancer
53. Rhome R, Singh A, Kechichian T, Drago M, Morace G, 11:775–791.
Luberto C, Del Poeta M. 2011. Surface localization 65. Bahn YS, Jung KW. 2013. Stress signaling pathways for
of glucosylceramide during Cryptococcus neoformans the pathogenicity of Cryptococcus. Eukaryot Cell 12:
infection allows targeting as a potential antifungal. 1564–1577.
PLoS One 6:e15572. 66. Hogan DA, Sundstrom P. 2009. The Ras/cAMP/PKA
54. Tafesse FG, Rashidfarrokhi A, Schmidt FI, Freinkman signaling pathway and virulence in Candida albicans.
E, Dougan S, Dougan M, Esteban A, Maruyama T, Future Microbiol 4:1263–1270.
Strijbis K, Ploegh HL. 2015. Disruption of sphingolipid 67. He B, Chen P, Chen SY, Vancura KL, Michaelis S,
biosynthesis blocks phagocytosis of Candida albicans. Powers S. 1991. RAM2, an essential gene of yeast, and
PLoS Pathog 11:e1005188. RAM1 encode the two polypeptide components of the
55. Tiede A, Bastisch I, Schubert J, Orlean P, Schmidt RE. farnesyltransferase that prenylates a-factor and Ras pro-
1999. Biosynthesis of glycosylphosphatidylinositols in teins. Proc Natl Acad Sci USA 88:11373–11377.
mammals and unicellular microbes. Biol Chem 380: 68. Vallim MA, Fernandes L, Alspaugh JA. 2004. The
503–523. RAM1 gene encoding a protein-farnesyltransferase
56. Richard M, Ibata-Ombetta S, Dromer F, Bordon-Pallier beta-subunit homologue is essential in Cryptococcus
F, Jouault T, Gaillardin C. 2002. Complete glycosyl- neoformans. Microbiology 150:1925–1935.
phosphatidylinositol anchors are required in Candida 69. Song JL, White TC. 2003. RAM2: an essential gene in
albicans for full morphogenesis, virulence and resistance
the prenylation pathway of Candida albicans. Micro-
to macrophages. Mol Microbiol 44:841–853. biology 149:249–259.
57. Yan J, Du T, Zhao W, Hartmann T, Lu H, Lü Y,
70. Fortwendel JR, Juvvadi PR, Rogg LE, Asfaw YG,
Ouyang H, Jiang X, Sun L, Jin C. 2013. Transcriptome
Burns KA, Randell SH, Steinbach WJ. 2012. Plasma
and biochemical analysis reveals that suppression of
membrane localization is required for RasA-mediated
GPI-anchor synthesis leads to autophagy and possible
polarized morphogenesis and virulence of Aspergillus
necroptosis in Aspergillus fumigatus. PLoS One 8:
fumigatus. Eukaryot Cell 11:966–977.
e59013.
58. Li H, Zhou H, Luo Y, Ouyang H, Hu H, Jin C. 2007. 71. Hast MA, Nichols CB, Armstrong SM, Kelly SM,
Glycosylphosphatidylinositol (GPI) anchor is required Hellinga HW, Alspaugh JA, Beese LS. 2011. Structures
of Cryptococcus neoformans protein farnesyltransferase
in Aspergillus fumigatus for morphogenesis and viru-
lence. Mol Microbiol 64:1014–1027. reveal strategies for developing inhibitors that target
fungal pathogens. J Biol Chem 286:35149–35162.
59. Romano J, Nimrod G, Ben-Tal N, Shadkchan Y,
Baruch K, Sharon H, Osherov N. 2006. Disruption of 72. Qiao J, Gao P, Jiang X, Fang H. 2013. In vitro anti-
the Aspergillus fumigatus ECM33 homologue results fungal activity of farnesyltransferase inhibitors against
in rapid conidial germination, antifungal resistance and clinical isolates of Aspergillus and Candida. Ann Clin
hypervirulence. Microbiology 152:1919–1928. Microbiol Antimicrob 12:37.
60. Tsukahara K, Hata K, Nakamoto K, Sagane K, 73. McGeady P, Logan DA, Wansley DL. 2002. A protein-
Watanabe NA, Kuromitsu J, Kai J, Tsuchiya M, Ohba farnesyl transferase inhibitor interferes with the serum-
F, Jigami Y, Yoshimatsu K, Nagasu T. 2003. Medicinal induced conversion of Candida albicans from a cellular
genetics approach towards identifying the molecular tar- yeast form to a filamentous form. FEMS Microbiol Lett
get of a novel inhibitor of fungal cell wall assembly. Mol 213:41–44.
Microbiol 48:1029–1042. 74. Mabanglo MF, Hast MA, Lubock NB, Hellinga HW,
61. Hata K, Horii T, Miyazaki M, Watanabe NA, Okubo Beese LS. 2014. Crystal structures of the fungal patho-
M, Sonoda J, Nakamoto K, Tanaka K, Shirotori S, gen Aspergillus fumigatus protein farnesyltransferase
Murai N, Inoue S, Matsukura M, Abe S, Yoshimatsu complexed with substrates and inhibitors reveal features
K, Asada M. 2011. Efficacy of oral E1210, a new for antifungal drug design. Protein Sci 23:289–301.
broad-spectrum antifungal with a novel mechanism of 75. Panepinto JC, Oliver BG, Fortwendel JR, Smith DL,
action, in murine models of candidiasis, aspergillosis, Askew DS, Rhodes JC. 2003. Deletion of the Aspergil-
and fusariosis. Antimicrob Agents Chemother 55:4543– lus fumigatus gene encoding the Ras-related protein
4551. RhbA reduces virulence in a model of invasive pulmo-
62. McLellan CA, Whitesell L, King OD, Lancaster AK, nary aspergillosis. Infect Immun 71:2819–2826.
Mazitschek R, Lindquist S. 2012. Inhibiting GPI anchor 76. Fox DS, Heitman J. 2002. Good fungi gone bad: the
biosynthesis in fungi stresses the endoplasmic reticulum corruption of calcineurin. BioEssays 24:894–903.
and enhances immunogenicity. ACS Chem Biol 7:1520– 77. Steinbach WJ, Reedy JL, Cramer RA Jr, Perfect JR,
1528. Heitman J. 2007. Harnessing calcineurin as a novel
63. Shen H, Chen SM, Liu W, Zhu F, He LJ, Zhang JD, anti-infective agent against invasive fungal infections.
Zhang SQ, Yan L, Xu Z, Xu GT, An MM, Jiang YY. Nat Rev Microbiol 5:418–430.
44. ANTIFUNGAL DRUGS: CURRENT ARMAMENTARIUM AND NEW AGENTS 919

78. Odom A, Muir S, Lim E, Toffaletti DL, Perfect J, formans by inhibition of a common target protein.
Heitman J. 1997. Calcineurin is required for virulence Antimicrob Agents Chemother 41:156–161.
of Cryptococcus neoformans. EMBO J 16:2576–2589. 92. Lugardon K, Chasserot-Golaz S, Kieffer AE, Maget-
79. Cruz MC, Goldstein AL, Blankenship JR, Del Poeta M, Dana R, Nullans G, Kieffer B, Aunis D, Metz-Boutigue
Davis D, Cardenas ME, Perfect JR, McCusker JH, MH. 2001. Structural and biological characterization of
Heitman J. 2002. Calcineurin is essential for survival chromofungin, the antifungal chromogranin A-(47-66)-
during membrane stress in Candida albicans. EMBO J derived peptide. J Biol Chem 276:35875–35882.
21:546–559. 93. Pearl LH, Prodromou C. 2006. Structure and mecha-
80. Wiederhold NP, Kontoyiannis DP, Prince RA, Lewis nism of the Hsp90 molecular chaperone machinery.
RE. 2005. Attenuation of the activity of caspofungin at Annu Rev Biochem 75:271–294.
high concentrations against Candida albicans: possible 94. Taipale M, Jarosz DF, Lindquist S. 2010. HSP90 at
role of cell wall integrity and calcineurin pathways. the hub of protein homeostasis: emerging mechanistic
Antimicrob Agents Chemother 49:5146–5148. insights. Nat Rev Mol Cell Biol 11:515–528.
81. Singh SD, Robbins N, Zaas AK, Schell WA, Perfect JR, 95. Queitsch C, Sangster TA, Lindquist S. 2002. Hsp90 as a
Cowen LE. 2009. Hsp90 governs echinocandin resis- capacitor of phenotypic variation. Nature 417:618–624.
tance in the pathogenic yeast Candida albicans via 96. Rutherford SL, Lindquist S. 1998. Hsp90 as a capacitor
calcineurin. PLoS Pathog 5:e1000532. for morphological evolution. Nature 396:336–342.
82. Cowen LE, Lindquist S. 2005. Hsp90 potentiates the 97. Jarosz DF, Lindquist S. 2010. Hsp90 and environmental
rapid evolution of new traits: drug resistance in diverse stress transform the adaptive value of natural genetic
fungi. Science 309:2185–2189. variation. Science 330:1820–1824.
83. Cowen LE, Carpenter AE, Matangkasombut O, Fink 98. Cowen LE. 2013. The fungal Achilles’ heel: targeting
GR, Lindquist S. 2006. Genetic architecture of Hsp90- Hsp90 to cripple fungal pathogens. Curr Opin Micro-
dependent drug resistance. Eukaryot Cell 5:2184–2188. biol 16:377–384.
84. Onyewu C, Blankenship JR, Del Poeta M, Heitman J.
99. Veri A, Cowen LE. 2014. Progress and prospects for
2003. Ergosterol biosynthesis inhibitors become fungi- targeting Hsp90 to treat fungal infections. Parasitology
cidal when combined with calcineurin inhibitors against
141:1127–1137.
Candida albicans, Candida glabrata, and Candida
krusei. Antimicrob Agents Chemother 47:956–964. 100. Lamoth F, Juvvadi PR, Steinbach WJ. 2016. Heat shock
protein 90 (Hsp90): a novel antifungal target against
85. Uppuluri P, Nett J, Heitman J, Andes D. 2008. Syner-
Aspergillus fumigatus. Crit Rev Microbiol 42:310–321.
gistic effect of calcineurin inhibitors and fluconazole
against Candida albicans biofilms. Antimicrob Agents 101. Cordeiro RA, Evangelista AJ, Serpa R, Marques FJ, de
Chemother 52:1127–1132. Melo CV, de Oliveira JS, Franco JS, de Alencar LP,
86. Kontoyiannis DP, Lewis RE, Osherov N, Albert ND, Bandeira TJ, Brilhante RS, Sidrim JJ, Rocha MF. 2016.
May GS. 2003. Combination of caspofungin with in- Inhibition of heat-shock protein 90 enhances the sus-
ceptibility to antifungals and reduces the virulence of
hibitors of the calcineurin pathway attenuates growth
in vitro in Aspergillus species. J Antimicrob Chemother Cryptococcus neoformans/Cryptococcus gattii species
51:313–316. complex. Microbiology 162:309–317.
87. Steinbach WJ, Schell WA, Blankenship JR, Onyewu C, 102. Lamoth F, Juvvadi PR, Fortwendel JR, Steinbach WJ.
Heitman J, Perfect JR. 2004. In vitro interactions be- 2012. Heat shock protein 90 is required for conidia-
tween antifungals and immunosuppressants against tion and cell wall integrity in Aspergillus fumigatus.
Aspergillus fumigatus. Antimicrob Agents Chemother Eukaryot Cell 11:1324–1332.
48:1664–1669. 103. Singh-Babak SD, Babak T, Diezmann S, Hill JA, Xie
88. Lamoth F, Juvvadi PR, Gehrke C, Steinbach WJ. JL, Chen YL, Poutanen SM, Rennie RP, Heitman J,
2013. In vitro activity of calcineurin and heat shock Cowen LE. 2012. Global analysis of the evolution and
protein 90 inhibitors against Aspergillus fumigatus mechanism of echinocandin resistance in Candida
azole- and echinocandin-resistant strains. Antimicrob glabrata. PLoS Pathog 8:e1002718.
Agents Chemother 57:1035–1039. 104. Lamoth F, Juvvadi PR, Gehrke C, Asfaw YG, Steinbach
89. Lee SC, Li A, Calo S, Inoue M, Tonthat NK, Bain JM, WJ. 2014. Transcriptional activation of heat shock pro-
Louw J, Shinohara ML, Erwig LP, Schumacher MA, tein 90 mediated via a proximal promoter region as
Ko DC, Heitman J. 2015. Calcineurin orchestrates di- trigger of caspofungin resistance in Aspergillus
morphic transitions, antifungal drug responses and host- fumigatus. J Infect Dis 209:473–481.
pathogen interactions of the pathogenic mucoralean 105. LaFayette SL, Collins C, Zaas AK, Schell WA, Betancourt-
fungus Mucor circinelloides. Mol Microbiol 97:844–865. Quiroz M, Gunatilaka AA, Perfect JR, Cowen LE.
90. Lee SC, Li A, Calo S, Heitman J. 2013. Calcineurin 2010. PKC signaling regulates drug resistance of the
plays key roles in the dimorphic transition and virulence fungal pathogen Candida albicans via circuitry com-
of the human pathogenic zygomycete Mucor circinel- prised of Mkc1, calcineurin, and Hsp90. PLoS Pathog
loides. PLoS Pathog 9:e1003625. 6:e1001069.
91. Odom A, Del Poeta M, Perfect J, Heitman J. 1997. The 106. Hill JA, O’Meara TR, Cowen LE. 2015. Fitness
immunosuppressant FK506 and its nonimmunosuppres- trade-offs associated with the evolution of resistance to
sive analog L-685,818 are toxic to Cryptococcus neo- antifungal drug combinations. Cell Rep 10:809–819.
920 FUNGI AND THE HUMAN HOST

107. McClellan AJ, Xia Y, Deutschbauer AM, Davis RW, 121. Wurtele H, Tsao S, Lépine G, Mullick A, Tremblay J,
Gerstein M, Frydman J. 2007. Diverse cellular functions Drogaris P, Lee EH, Thibault P, Verreault A, Raymond
of the Hsp90 molecular chaperone uncovered using sys- M. 2010. Modulation of histone H3 lysine 56 acetyla-
tems approaches. Cell 131:121–135. tion as an antifungal therapeutic strategy. Nat Med 16:
108. Zhao R, Davey M, Hsu YC, Kaplanek P, Tong A, 774–780.
Parsons AB, Krogan N, Cagney G, Mai D, Greenblatt J, 122. Pfaller MA, Messer SA, Georgopapadakou N, Martell
Boone C, Emili A, Houry WA. 2005. Navigating the LA, Besterman JM, Diekema DJ. 2009. Activity of
chaperone network: an integrative map of physical and MGCD290, a Hos2 histone deacetylase inhibitor,
genetic interactions mediated by the hsp90 chaperone. in combination with azole antifungals against opportu-
Cell 120:715–727. nistic fungal pathogens. J Clin Microbiol 47:3797–
109. Diezmann S, Michaut M, Shapiro RS, Bader GD, 3804.
Cowen LE. 2012. Mapping the Hsp90 genetic interac- 123. Pfaller MA, Rhomberg PR, Messer SA, Castanheira M.
tion network in Candida albicans reveals environmental 2015. In vitro activity of a Hos2 deacetylase inhibitor,
contingency and rewired circuitry. PLoS Genet 8: MGCD290, in combination with echinocandins against
e1002562. echinocandin-resistant Candida species. Diagn Micro-
110. Shapiro RS, Uppuluri P, Zaas AK, Collins C, Senn H, biol Infect Dis 81:259–263.
Perfect JR, Heitman J, Cowen LE. 2009. Hsp90 orches- 124. Besterman J, Nguyen DT, Ste-Croix H. 2012.
trates temperature-dependent Candida albicans morpho- MGCD290, an oral fungal Hos2 inhibitor, enhances the
genesis via Ras1-PKA signaling. Curr Biol 19:621–629. antifungal properties of fluconazole following multiple-
111. Shapiro RS, Zaas AK, Betancourt-Quiroz M, Perfect or single-dose oral administration in pre-and post-
JR, Cowen LE. 2012. The Hsp90 co-chaperone Sgt1 infection settings. MethylGene, Inc. Abstr ICAAC/ICC
governs Candida albicans morphogenesis and drug re- 2015, San Diego, abstr M-1711.
sistance. PLoS One 7:e44734. 125. Lamoth F, Juvvadi PR, Soderblom EJ, Moseley MA,
112. Shapiro RS, Cowen L. 2010. Coupling temperature Asfaw YG, Steinbach WJ. 2014. Identification of a
sensing and development: Hsp90 regulates morphoge- key lysine residue in heat shock protein 90 required for
netic signalling in Candida albicans. Virulence 1:45–48. azole and echinocandin resistance in Aspergillus fumi-
113. Senn H, Shapiro RS, Cowen LE. 2012. Cdc28 provides gatus. Antimicrob Agents Chemother 58:1889–1896.
a molecular link between Hsp90, morphogenesis, and 126. Sellam A, Askew C, Epp E, Lavoie H, Whiteway M,
cell cycle progression in Candida albicans. Mol Biol Nantel A. 2009. Genome-wide mapping of the coacti-
Cell 23:268–283. vator Ada2p yields insight into the functional roles of
114. Shapiro RS, Sellam A, Tebbji F, Whiteway M, Nantel SAGA/ADA complex in Candida albicans. Mol Biol
A, Cowen LE. 2012. Pho85, Pcl1, and Hms1 signaling Cell 20:2389–2400.
governs Candida albicans morphogenesis induced by 127. Lopes da Rosa J, Boyartchuk VL, Zhu LJ, Kaufman PD.
high temperature or Hsp90 compromise. Curr Biol 22: 2010. Histone acetyltransferase Rtt109 is required for
461–470. Candida albicans pathogenesis. Proc Natl Acad Sci USA
115. Cowen LE, Singh SD, Köhler JR, Collins C, Zaas AK, 107:1594–1599.
Schell WA, Aziz H, Mylonakis E, Perfect JR, Whitesell 128. Schmelzle T, Hall MN. 2000. TOR, a central controller
L, Lindquist S. 2009. Harnessing Hsp90 function as of cell growth. Cell 103:253–262.
a powerful, broadly effective therapeutic strategy for 129. De Virgilio C, Loewith R. 2006. Cell growth control:
fungal infectious disease. Proc Natl Acad Sci USA 106: little eukaryotes make big contributions. Oncogene 25:
2818–2823. 6392–6415.
116. Robbins N, Uppuluri P, Nett J, Rajendran R, Ramage 130. Wullschleger S, Loewith R, Hall MN. 2006. TOR sig-
G, Lopez-Ribot JL, Andes D, Cowen LE. 2011. Hsp90 naling in growth and metabolism. Cell 124:471–484.
governs dispersion and drug resistance of fungal 131. Vézina C, Kudelski A, Sehgal SN. 1975. Rapamycin
biofilms. PLoS Pathog 7:e1002257. (AY-22,989), a new antifungal antibiotic. I. Taxonomy
117. Shahbazian MD, Grunstein M. 2007. Functions of of the producing streptomycete and isolation of the ac-
site-specific histone acetylation and deacetylation. Annu tive principle. J Antibiot (Tokyo) 28:721–726.
Rev Biochem 76:75–100. 132. Heitman J, Movva NR, Hall MN. 1991. Targets for cell
118. Yang XJ, Seto E. 2008. The Rpd3/Hda1 family of lysine cycle arrest by the immunosuppressant rapamycin in
deacetylases: from bacteria and yeast to mice and men. yeast. Science 253:905–909.
Nat Rev Mol Cell Biol 9:206–218. 133. Cruz MC, Cavallo LM, Görlach JM, Cox G, Perfect JR,
119. Smith WL, Edlind TD. 2002. Histone deacetylase inhib- Cardenas ME, Heitman J. 1999. Rapamycin antifun-
itors enhance Candida albicans sensitivity to azoles and gal action is mediated via conserved complexes with
related antifungals: correlation with reduction in CDR FKBP12 and TOR kinase homologs in Cryptococcus
and ERG upregulation. Antimicrob Agents Chemother neoformans. Mol Cell Biol 19:4101–4112.
46:3532–3539. 134. Wong GK, Griffith S, Kojima I, Demain AL. 1998.
120. Robbins N, Leach MD, Cowen LE. 2012. Lysine Antifungal activities of rapamycin and its deriva-
deacetylases Hda1 and Rpd3 regulate Hsp90 function tives, prolylrapamycin, 32-desmethylrapamycin, and
thereby governing fungal drug resistance. Cell Rep 2: 32-desmethoxyrapamycin. J Antibiot (Tokyo) 51:487–
878–888. 491.
44. ANTIFUNGAL DRUGS: CURRENT ARMAMENTARIUM AND NEW AGENTS 921

135. Robbins N, Collins C, Morhayim J, Cowen LE. 2010. 150. Roemer T, Boone C. 2013. Systems-level antimicrobial
Metabolic control of antifungal drug resistance. Fungal drug and drug synergy discovery. Nat Chem Biol 9:
Genet Biol 47:81–93. 222–231.
136. Cruz MC, Goldstein AL, Blankenship J, Del Poeta M, 151. Sandovsky-Losica H, Shwartzman R, Lahat Y, Segal E.
Perfect JR, McCusker JH, Bennani YL, Cardenas ME, 2008. Antifungal activity against Candida albicans
Heitman J. 2001. Rapamycin and less immunosuppres- of nikkomycin Z in combination with caspofungin,
sive analogs are toxic to Candida albicans and Crypto- voriconazole or amphotericin B. J Antimicrob Chemo-
coccus neoformans via FKBP12-dependent inhibition of ther 62:635–637.
TOR. Antimicrob Agents Chemother 45:3162–3170. 152. Verwer PE, van Duijn ML, Tavakol M, Bakker-
137. Rock FL, Mao W, Yaremchuk A, Tukalo M, Crépin T, Woudenberg IA, van de Sande WW. 2012. Reshuffling
Zhou H, Zhang YK, Hernandez V, Akama T, Baker of Aspergillus fumigatus cell wall components chitin
SJ, Plattner JJ, Shapiro L, Martinis SA, Benkovic SJ, and β-glucan under the influence of caspofungin or
Cusack S, Alley MR. 2007. An antifungal agent inhibits nikkomycin Z alone or in combination. Antimicrob
an aminoacyl-tRNA synthetase by trapping tRNA in Agents Chemother 56:1595–1598.
the editing site. Science 316:1759–1761. 153. Wildenhain J, Spitzer M, Dolma S, Jarvik N, White R,
138. Seiradake E, Mao W, Hernandez V, Baker SJ, Plattner Roy M, Griffiths E, Bellows DS, Wright GD, Tyers M.
JJ, Alley MR, Cusack S. 2009. Crystal structures of the 2015. Prediction of synergism from chemical-genetic in-
human and fungal cytosolic leucyl-tRNA synthetase teractions by machine learning. Cell Syst 1:383–395.
editing domains: a structural basis for the rational de- 154. Epp E, Vanier G, Harcus D, Lee AY, Jansen G, Hallett
sign of antifungal benzoxaboroles. J Mol Biol 390: M, Sheppard DC, Thomas DY, Munro CA, Mullick A,
196–207. Whiteway M. 2010. Reverse genetics in Candida
139. Wright GD. 2015. Solving the antibiotic crisis. ACS albicans predicts ARF cycling is essential for drug resis-
Infect Dis 1:80–84. tance and virulence. PLoS Pathog 6:e1000753.
140. Brown ED, Wright GD. 2016. Antibacterial drug dis- 155. Zhai B, Wu C, Wang L, Sachs MS, Lin X. 2012. The
covery in the resistance era. Nature 529:336–343. antidepressant sertraline provides a promising therapeu-
141. Zimmermann GR, Lehár J, Keith CT. 2007. Multi- tic option for neurotropic cryptococcal infections.
target therapeutics: when the whole is greater than the Antimicrob Agents Chemother 56:3758–3766.
sum of the parts. Drug Discov Today 12:34–42. 156. Butts A, DiDone L, Koselny K, Baxter BK, Chabrier-
142. Hill JA, Cowen LE. 2015. Using combination therapy to Rosello Y, Wellington M, Krysan DJ. 2013. A repur-
thwart drug resistance. Future Microbiol 10:1719–1726. posing approach identifies off-patent drugs with fungicidal
cryptococcal activity, a common structural chemotype,
143. Baym M, Stone LK, Kishony R. 2016. Multidrug evolu- and pharmacological properties relevant to the treatment
tionary strategies to reverse antibiotic resistance. Sci-
of cryptococcosis. Eukaryot Cell 12:278–287.
ence 351:aad3292.
157. Robbins N, Spitzer M, Yu T, Cerone RP, Averette AK,
144. Bock C, Lengauer T. 2012. Managing drug resistance Bahn YS, Heitman J, Sheppard DC, Tyers M, Wright
in cancer: lessons from HIV therapy. Nat Rev Cancer GD. 2015. An antifungal combination matrix identifies
12:494–501. a rich pool of adjuvant molecules that enhance drug
145. Zumla A, Hafner R, Lienhardt C, Hoelscher M, Nunn activity against diverse fungal pathogens. Cell Rep 13:
A. 2012. Advancing the development of tuberculosis 1481–1492.
therapy. Nat Rev Drug Discov 11:171–172. 158. Butts A, Koselny K, Chabrier-Roselló Y, Semighini CP,
146. Eastman RT, Fidock DA. 2009. Artemisinin-based com- Brown JC, Wang X, Annadurai S, DiDone L, Tabroff J,
bination therapies: a vital tool in efforts to eliminate Childers WE Jr, Abou-Gharbia M, Wellington M,
malaria. Nat Rev Microbiol 7:864–874. Cardenas ME, Madhani HD, Heitman J, Krysan DJ. 2014.
147. Tong AH, Lesage G, Bader GD, Ding H, Xu H, Xin X, Estrogen receptor antagonists are anti-cryptococcal
Young J, Berriz GF, Brost RL, Chang M, Chen Y, agents that directly bind EF hand proteins and synergize
Cheng X, Chua G, Friesen H, Goldberg DS, Haynes J, with fluconazole in vivo. MBio 5:e00765–e13.
Humphries C, He G, Hussein S, Ke L, Krogan N, Li Z, 159. Nishikawa JL, Boeszoermenyi A, Vale-Silva LA,
Levinson JN, Lu H, Ménard P, Munyana C, Parsons Torelli R, Posteraro B, Sohn YJ, Ji F, Gelev V, Sanglard
AB, Ryan O, Tonikian R, Roberts T, Sdicu AM, D, Sanguinetti M, Sadreyev RI, Mukherjee G,
Shapiro J, Sheikh B, Suter B, Wong SL, Zhang LV, Zhu Bhyravabhotla J, Buhrlage SJ, Gray NS, Wagner G,
H, Burd CG, Munro S, Sander C, Rine J, Greenblatt J, Näär AM, Arthanari H. 2016. Inhibiting fungal multi-
Peter M, Bretscher A, Bell G, Roth FP, Brown GW, drug resistance by disrupting an activator-mediator in-
Andrews B, Bussey H, Boone C. 2004. Global mapping teraction. Nature 530:485–489.
of the yeast genetic interaction network. Science 303: 160. Borisy AA, Elliott PJ, Hurst NW, Lee MS, Lehar J,
808–813. Price ER, Serbedzija G, Zimmermann GR, Foley MA,
148. Costanzo M, et al. 2010. The genetic landscape of a Stockwell BR, Keith CT. 2003. Systematic discovery of
cell. Science 327:425–431. multicomponent therapeutics. Proc Natl Acad Sci USA
149. Winzeler EA, et al. 1999. Functional characterization of 100:7977–7982.
the S. cerevisiae genome by gene deletion and parallel 161. Zhang L, Yan K, Zhang Y, Huang R, Bian J, Zheng C,
analysis. Science 285:901–906. Sun H, Chen Z, Sun N, An R, Min F, Zhao W, Zhuo Y,
922 FUNGI AND THE HUMAN HOST

You J, Song Y, Yu Z, Liu Z, Yang K, Gao H, Dai H, WS, Brown GW, Schimmer AD, Bankaitis VA, Nislow
Zhang X, Wang J, Fu C, Pei G, Liu J, Zhang S, C, Bader GD, Giaever G. 2014. Mapping the cellular
Goodfellow M, Jiang Y, Kuai J, Zhou G, Chen X. response to small molecules using chemogenomic fitness
2007. High-throughput synergy screening identifies mi- signatures. Science 344:208–211.
crobial metabolites as combination agents for the treat- 173. Roemer T, Jiang B, Davison J, Ketela T, Veillette K,
ment of fungal infections. Proc Natl Acad Sci USA 104: Breton A, Tandia F, Linteau A, Sillaots S, Marta C,
4606–4611. Martel N, Veronneau S, Lemieux S, Kauffman S, Becker
162. Lehár J, Zimmermann GR, Krueger AS, Molnar RA, J, Storms R, Boone C, Bussey H. 2003. Large-scale
Ledell JT, Heilbut AM, Short GF III, Giusti LC, Nolan essential gene identification in Candida albicans and
GP, Magid OA, Lee MS, Borisy AA, Stockwell BR, applications to antifungal drug discovery. Mol Micro-
Keith CT. 2007. Chemical combination effects predict biol 50:167–181.
connectivity in biological systems. Mol Syst Biol 3:80. 174. Xu D, Jiang B, Ketela T, Lemieux S, Veillette K, Martel
163. Jansen G, Lee AY, Epp E, Fredette A, Surprenant J, N, Davison J, Sillaots S, Trosok S, Bachewich C, Bussey
Harcus D, Scott M, Tan E, Nishimura T, Whiteway M, H, Youngman P, Roemer T. 2007. Genome-wide fitness
Hallett M, Thomas DY. 2009. Chemogenomic profiling test and mechanism-of-action studies of inhibitory
predicts antifungal synergies. Mol Syst Biol 5:338. compounds in Candida albicans. PLoS Pathog 3:e92.
164. Lo YC, Senese S, Li CM, Hu Q, Huang Y, Damoiseaux 175. Brown JC, Nelson J, VanderSluis B, Deshpande R,
R, Torres JZ. 2015. Large-scale chemical similarity Butts A, Kagan S, Polacheck I, Krysan DJ, Myers CL,
networks for target profiling of compounds identified in Madhani HD. 2014. Unraveling the biology of a fungal
cell-based chemical screens. PLOS Comput Biol 11: meningitis pathogen using chemical genetics. Cell 159:
e1004153. 1168–1187.
165. Spitzer M, Griffiths E, Blakely KM, Wildenhain J, Ejim 176. O’Meara TR, Veri AO, Ketela T, Jiang B, Roemer T,
L, Rossi L, De Pascale G, Curak J, Brown E, Tyers M, Cowen LE. 2015. Global analysis of fungal morphology
Wright GD. 2011. Cross-species discovery of syncretic exposes mechanisms of host cell escape. Nat Commun
drug combinations that potentiate the antifungal flu- 6:6741.
conazole. Mol Syst Biol 7:499. 177. Huang Z, Chen K, Zhang J, Li Y, Wang H, Cui D, Tang
166. Roemer T, Davies J, Giaever G, Nislow C. 2011. Bugs, J, Liu Y, Shi X, Li W, Liu D, Chen R, Sucgang RS, Pan
drugs and chemical genomics. Nat Chem Biol 8:46–56. X. 2013. A functional variomics tool for discovering
167. Giaever G, Flaherty P, Kumm J, Proctor M, Nislow C, drug-resistance genes and drug targets. Cell Reports 3:
Jaramillo DF, Chu AM, Jordan MI, Arkin AP, Davis 577–585.
RW. 2004. Chemogenomic profiling: identifying the 178. Pries V, Cotesta S, Riedl R, Aust T, Schuierer S, Tao J,
functional interactions of small molecules in yeast. Proc Filipuzzi I, Hoepfner D. 2016. Advantages and chal-
Natl Acad Sci USA 101:793–798. lenges of phenotypic screens: the identification of two
168. Giaever G, Shoemaker DD, Jones TW, Liang H, novel antifungal geranylgeranyltransferase I inhibitors.
Winzeler EA, Astromoff A, Davis RW. 1999. Genomic J Biomol Screen 21:306–315.
profiling of drug sensitivities via induced haploin- 179. Baltz RH. 2009. Daptomycin: mechanisms of action
sufficiency. Nat Genet 21:278–283. and resistance, and biosynthetic engineering. Curr Opin
169. Giaever G, et al. 2002. Functional profiling of the Chem Biol 13:144–151.
Saccharomyces cerevisiae genome. Nature 418:387– 180. Anniballi F, Lonati D, Fiore A, Auricchio B, De Medici
391. D, Locatelli CA. 2014. New targets in the search for
preventive and therapeutic agents for botulism. Expert
170. Lee W, St Onge RP, Proctor M, Flaherty P, Jordan MI,
Rev Anti Infect Ther 12:1075–1086.
Arkin AP, Davis RW, Nislow C, Giaever G. 2005.
Genome-wide requirements for resistance to functionally 181. Clerici M, Piconi S, Trabattoni D. 1999. Vaccine strate-
distinct DNA-damaging agents. PLoS Genet 1:e24. gies for infectious diseases. Expert Opin Investig Drugs
8:95–106.
171. Hillenmeyer ME, Fung E, Wildenhain J, Pierce SE,
Hoon S, Lee W, Proctor M, St Onge RP, Tyers M, 182. Clatworthy AE, Pierson E, Hung DT. 2007. Targeting
Koller D, Altman RB, Davis RW, Nislow C, Giaever G. virulence: a new paradigm for antimicrobial therapy.
2008. The chemical genomic portrait of yeast: uncover- Nat Chem Biol 3:541–548.
ing a phenotype for all genes. Science 320:362–365. 183. Gauwerky K, Borelli C, Korting HC. 2009. Targeting
172. Lee AY, St Onge RP, Proctor MJ, Wallace IM, Nile AH, virulence: a new paradigm for antifungals. Drug Discov
Spagnuolo PA, Jitkova Y, Gronda M, Wu Y, Kim MK, Today 14:214–222.
Cheung-Ong K, Torres NP, Spear ED, Han MK, 184. Denning DW, Bromley MJ. 2015. How to bolster the
Schlecht U, Suresh S, Duby G, Heisler LE, Surendra A, antifungal pipeline. Science 347:1414–1416.
Fung E, Urbanus ML, Gebbia M, Lissina E, Miranda 185. Cowen LE. 2008. The evolution of fungal drug resis-
M, Chiang JH, Aparicio AM, Zeghouf M, Davis RW, tance: modulating the trajectory from genotype to phe-
Cherfils J, Boutry M, Kaiser CA, Cummins CL, Trimble notype. Nat Rev Microbiol 6:187–198.

You might also like