Antifungi

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 11

Biochemical Pharmacology xxx (2016) xxx–xxx

Contents lists available at ScienceDirect

Biochemical Pharmacology
journal homepage: www.elsevier.com/locate/biochempharm

Review

Antifungals
Sonia Campoy, José L. Adrio ⇑
Neol Biosolutions, S.A., c/Avicena, 4, Parque Tecnológico de la Salud, 18016 Granada, Spain

a r t i c l e i n f o a b s t r a c t

Article history: The need for new antifungal agents is undeniable. Current therapeutic choices for the treatment of
Received 25 September 2016 invasive fungal infections are limited to three classes of drugs. Most used antifungal agents are not com-
Accepted 18 November 2016 pletely effective due to the development of resistance, host toxicity and undesirable side effects that limit
Available online xxxx
their use in medical practice. Invasive fungal infections have significantly increased over the last decades
and the mortality rates remain unacceptably high. More threatening, new resistance patterns have been
Keywords: observed including simultaneous resistance to different antifungal classes.
Antifungals
In the last years, deeper insights into the molecular mechanisms for fungal resistance and virulence
Mycosis
Fungal resistance
have yielded some new potential targets for antifungal therapeutics. Chemical genomics-based screen-
Candida ings, high throughput screenings of natural products and repurposing of approved drugs are some of
Aspergillus the approaches being followed for the discovery of new antifungal molecules. However, despite the
Azole emerging need for effective antifungal agents, the current pipeline contains only a few promising
Polyenes molecules, with novel modes of action, in early clinical development stages.
Echinocandins ! 2016 Elsevier Inc. All rights reserved.
Pyrimidine analogs
Aspergillosis
Candidiasis

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
2. Classification of antifungal agents. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
2.1. Inhibitors of ergosterol biosynthesis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
2.1.1. Azoles . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
2.1.2. Allylamines . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
2.1.3. Morpholines . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
2.2. Fungal membrane disruptors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
2.3. Fungal cell wall synthesis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
2.3.1. Inhibitors b-glucan synthesis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
2.3.2. Chitin synthesis inhibitors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
2.4. Sphingolipids biosynthesis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
2.5. Nucleic acid synthesis inhibitors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
2.6. Protein biosynthesis inhibitors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
2.7. Microtubules biosynthesis inhibitors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
3. Strategies to develop new antifungal compounds . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
4. Mechanisms of antifungal resistance . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
5. Antifungal pipeline. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
6. Conclusions. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00

⇑ Corresponding author.
E-mail address: jladrio@neolbio.com (J.L. Adrio).

http://dx.doi.org/10.1016/j.bcp.2016.11.019
0006-2952/! 2016 Elsevier Inc. All rights reserved.

Please cite this article in press as: S. Campoy, J.L. Adrio, Antifungals, Biochem. Pharmacol. (2016), http://dx.doi.org/10.1016/j.bcp.2016.11.019
2 S. Campoy, J.L. Adrio / Biochemical Pharmacology xxx (2016) xxx–xxx

1. Introduction or intravenously for the treatment of fungal infections [6,9]. A fifth


class, allylamines, is also existing although compounds of this class
Invasive fungal infections represent a continuous and serious are used only for treating superficial dermathophytic infections.
threat to human health and they are associated with at least 1.5 However, these antifungals have various drawbacks in terms of
million deaths worldwide each year [1,2]. Estimates in the litera- toxicity, spectrum of activity, safety and pharmacokinetic proper-
ture support a 30–40% mortality for invasive candidiasis, 20–30% ties [1,10]. The emergence of strains resistant to the current anti-
for disseminated cryptococcosis and a similar percentage for inva- fungal agents has led great efforts to develop new drugs with
sive aspergillosis [3,4]. Such infections are very common in different mechanisms of action that target the biosynthesis of fun-
immunocompromised patients as a result of aggressive therapies gal proteins, lipids and cell wall [10].
(e.g. anticancer chemotherapy, long-term corticosteroids treat-
ment, or organ transplant) and immunosuppressive infections such
2. Classification of antifungal agents
as HIV/AIDS. About 90% of these deaths are caused by species
belonging to genera of fungi Candida, Aspergillus, Cryptococcus,
Based on their targets for antifungal therapy (Fig. 1) antifungal
Pneumocystis, Mucor and Rhizopus [5]. However, new and emerging
agents can be classified in the following groups:
fungal pathogen species belonging to Zygomycetes, Fusarium or Sce-
dosporium have becoming increasingly important as etiological
agents of invasive mycoses [6]. 2.1. Inhibitors of ergosterol biosynthesis
Fungi also produced superficial infections (involving the skin
and mucosal surfaces) which have greater incidence than the inva- Ergosterol is the major component of the fungal cell membrane
sive infections diminishing the quality of life of affected individu- and contributes to a variety of cellular functions such as fluidity
als. Superficial mycoses are caused by Malasseria globose and M. and integrity of the membrane and the proper function of
furfur. Cutaneous and subcutaneous mycoses that affect kera- membrane-bound enzymes.
tinized structures are caused by dermatophyte genera like Tri-
chophyton, Epidermophyton and Microsporum [7]. Mucosal 2.1.1. Azoles
infections are mostly caused by opportunistic yeasts being those Azoles are by far the most common antifungal drugs in clinical
belonging to the Candida genus the most frequent by far. use. They are highly used in the treatment and the prevention of
In comparison with the development of new antibacterial mycoses due to their broad spectrum activity. Azoles inhibit the
drugs, antifungal drug development is more challenging because cytochrome P450-dependent enzyme 14a-lanosterol demethylase
fungi are eukaryotes and many potential targets for therapy are (CYP51) encoded by the ERG11 gene that converts lanosterol to
also found in humans with substantial host toxicity risk [8,9]. ergosterol (Fig. 1) in the cell membrane inhibiting fungal growth
Currently, four classes of antifungal agents (azoles, echinocan- and replication [11–13]. This enzyme contains an iron protopor-
dins, polyenes and pyrimidine analogs) are used orally, topically phyrin unit at its active site. Azoles bind to the iron of the

Fig. 1. Antifungal agents targets.

Please cite this article in press as: S. Campoy, J.L. Adrio, Antifungals, Biochem. Pharmacol. (2016), http://dx.doi.org/10.1016/j.bcp.2016.11.019
S. Campoy, J.L. Adrio / Biochemical Pharmacology xxx (2016) xxx–xxx 3

porphyrin and cause the blockade of the fungal ergosterol biosyn- azole with the broadest activity and was approved by FDA for pro-
thesis pathway resulting in the accumulation of 14-methylated phylaxis against invasive Aspergillus and Candida infections.
sterols [14]. The exact conformation of the active site differs Efinaconazole (Fig. 2 8) is a topical antifungal solution active
between fungal species and among the many mammalian P450 against dermatophytes and nondermatophytes, effective in treat-
mono-oxygenases [15]. The nature of the interaction between each ment of onychomycosis, as well as Candida spp. It was approved
azole molecule and each kind of P450 determines their antifungal by FDA in 2014 for the treatment of fungal nail infections [25].
characteristics and side effects. Isavuconazole is the most recently approved triazole in the US
The azoles are cyclic organic molecules that can be classified and the EU for the oral and intravenous treatment of invasive
into two groups: imidazoles and triazoles. Imidazoles (clotrima- aspergillosis and mucormycosis [25]. It has some advantages com-
zole, miconazole and ketoconazole) (Fig. 2 1–3) were the first pared with other approved azoles as it has expanded activity that
developed azoles [16,17]. However, because of their high toxicity, includes Zygomycetes such as Rhizopus, Mucor and Cunninghamella
severe side effects and numerous interactions with other drugs, species. Additionally, its intravenous preparation lacks cyclodex-
they were replaced by the triazoles. The first generation triazoles trin that is associated with nephrotoxicity in patients.
(Itraconazole and Fluconazole) exhibit a broader antifungal activity
spectrum as compared to the imidazoles and have significant 2.1.2. Allylamines
improved safety profiles. Fluconazole (Fig. 2 4) is active against These synthetic fungicidal agents block ergosterol biosynthesis
Candida species, Cryptococcus neoformans, Histoplasma, Blastomyces as they are reversible, non-competitive inhibitors of squalene
and Coccidioides species, but lacks activity against molds [18,19]. epoxidase (ERG1). This enzyme catalyzes the conversion of squa-
Itraconazole (Fig. 2 5) shows a broader spectrum of activity being lene into 2,3-squalene epoxide. The inhibition of this enzymatic
active against yeasts and Aspergillus species [5,20]. However, both activity leads to squalene accumulation (Fig. 1) [5,20] that may
have certain clinical limitations as they are ineffective against increase permeability leading to disruption of cellular organiza-
some emerging pathogens as Scedosporium, Fusarium and Muco- tion. Important members of this group include terbinafine and naf-
rales [6,7]. Increase in azole resistance is mainly due to its fungi- tifine [26]. Terbinafine (Fig. 2 9) was isolated from cultures of
static nature instead of fungicidal [16]. Streptomyces sp. KH-F12. It is active against Aspergillus, Fusarium
To solve these problems, a second generation of triazoles was and other filamentous fungi [12,20]. It is widely use for the treat-
developed. Voriconazole (Fig. 2 6) and Posaconazole (Fig. 2 7) were ment of nail infections. Naftifine (Fig. 2 10) is active against Tri-
approved by the US Food and Drug Administration (FDA) in 2002 chophyton, Epidermophyton and Microsporum. Fungistatic activity
and 2006, respectively [21–24]. They are considered fungicidal against Candida species [27,28].
[25] and have a broad spectrum of activity including Fusarium, Sce-
dosporium, Zygomycetes and Cryptococcus neoformans. Voriconazole 2.1.3. Morpholines
became the first-line antifungal drug for the treatment of invasive The amorolfine (Fig. 2 11) is a synthetic water soluble morpho-
aspergillosis due to A. fumigatus since its activity is superior to line derivative that inhibits two enzymes involved in ergosterol
many other antifungals for this infection [1]. Posaconazole is the biosynthesis, D7–D8 isomerase (ERG2) and the D14-reductase

Fig. 2. Antifungal agents structure. 1 Clotrimazole, 2 Miconazole, 3 Ketoconazole, 4 Fluconazole, 5 Itraconazole, 6 Voriconazole, 7 Posaconazole, 8 Efinaconazole, 9
Terbinafine, 10 Naftifine, 11 Amorolfine, 12 Nystatin, 13 Natamycin, 14 Amphotericin B, 15 Echinocandins, 16 Nikkomycin, 17 Polyoxins, 18 Aureobasidin A 19 Flucytosine, 20
Tavaborole, 21 Cispentacin, 22 Icofungipen, 23 Sordarins, 24 Griseofulvin.

Please cite this article in press as: S. Campoy, J.L. Adrio, Antifungals, Biochem. Pharmacol. (2016), http://dx.doi.org/10.1016/j.bcp.2016.11.019
4 S. Campoy, J.L. Adrio / Biochemical Pharmacology xxx (2016) xxx–xxx

(ERG24) (Fig. 1). Amorolfine is used in the topical treatment of nail act as noncompetitive inhibitors of b-(1,3)-D-glucan synthase
infections [29] and it has both fungistatic and fungicidal activity enzyme complex, specifically targeting the FsK1 subunit which
in vitro. leads to disruption of the structure of growing cell walls, resulting
in osmotic instability and fungal cells death (Fig. 1). Echinocandins
2.2. Fungal membrane disruptors are semisynthetic lipopeptides derived from fungal natural prod-
ucts. Caspofungin is derived from pneumocandin B produced by
Polyenes are macrocyclic organic molecules known as macro- Glarea lozoyensis, micafungin is a semisynthetic product of
lides. Most of them consist of a 20–40 carbon macrolactone ring echinocandin B produced by Aspergillus nidulans var. echinulatus,
conjugated with a d-mycosimine group [30]. Due to their amphi- and anidulafungin is produced by chemical modification of a fer-
philic structure, these molecules bind to the lipid bilayer and form mentation product of Coleophoma empetri [53,54]. The basic struc-
a complex with the ergosterol producing pores. Pore formation ture of echinocandins consists in a cyclic hexapeptide with
promotes disruption of the cell membrane, the leakage of the cyto- different side chains at position R5 (Fig. 2 15) which are responsi-
plasmic contents and oxidative damage result in fungal cell death ble for their antifungal activity [55,56]. At such position, caspo-
[31–33]. Recently, it has been reported that polyenes (ampho- fungin has a fatty acid, micafungin has a complex aromatic
tericin B) bind ergosterol and form an extramembranous fungicidal substituent and anidulafungin has an alkoxytriphenyl group.
sterol sponge destabilizing membrane function [34]. Polyenes These drugs are poorly absorbed in the gastrointestinal tract
were the first antifungal drugs for clinical use. They are fungicidal because of their high molecular weights. Echinocandins show very
and have the broadest spectrum of activity compared to any other good safety profiles and their toxicity is very low due to their
antifungal molecules. Nystatin, natamycin and amphotericin B are unique target, that is absent in mammalian cells and interactions
the only three polyenes in clinical use [35–37]. They are natural with other drugs are minimal [22]. However, they have a short
products and were isolated from the cultivation broths of Strepto- half-life so they must be administered once-daily intravenously
myces noursei, S. natalensis and S. nodosum, respectively. limiting their use to the hospital [57].
Nystatin (Fig. 2 12) and natamycin (Fig. 2 13) are active against Caspofungin, micafungin and anidulafungin have fungicidal
Cryptococcus, Candida, Aspergillus and Fusarium. Nystatin is used for activity both in vitro and in vivo against many strains of Candida
the treatment of cutaneous, vaginal and esophageal candidiasis, and fungistatic activity against Aspergillus spp. Micafungin and
and natamycin can be used for the treatment of fungal keratosis anidulafungin were licensed for the treatment of invasive candidi-
or corneal infections [38]. Amphotericin B (Fig. 2 14) is active asis and esophageal candidiasis; caspofungin was also approved
against most yeasts and filamentous fungi. It is recommended for for invasive aspergillosis [19,58–60].
the treatment of infections caused by Candida, Aspergillus, Fusar- Efforts to obtain new echinocandins are relevant since these
ium, Mucor, Scedosporium and Cryptococcus among others [39,40]. compounds show broad-spectrum antifungal activity and a possi-
Polyenes have a lower but non-negligible affinity for choles- ble use in combination with azoles or amphotericin B could
terol. This slight affinity for cholesterol explains the high toxicity enhance their action against fungal pathogens without causing
associated with these antifungals and is responsible for numerous the development of resistant strains [57].
side effects [41]. Nystatin and natamycin are only used as topical
agents due to their low absorption in the gut and their high toxic- 2.3.2. Chitin synthesis inhibitors
ity. Amphotericin B is the most used polyene for the treatment of Chitin is a minor component of the cell wall (3%) which consists
systemic infections. Due to its hydrophobicity and poor absorption of a linear homopolymer of b-(1,4)-linked N-acetylglucosamine
through the gastrointestinal tract, it is administered intravenously covalently linked to b-(1,3)-D-glucan [50,59,61]. The synthesis of
which causes adverse effects in kidneys and liver [42]. chitin is mediated by several enzymes named chitin synthases.
In recent years, the main efforts have been focused on finding Chitin provides structural integrity and when its synthesis is dis-
new molecules that minimize the toxicity of these three com- rupted, the cell wall becomes osmotically unstable. Because chitin
pounds [43]. New semi-synthetic polyenes have been developed is absent in human cells, it is considered an excellent target for
showing improved water-solubility, lower hemotoxicity than antifungal agents. There most widely studied chitin synthase inhi-
amphotericin B, superior selectivity toward ergosterol-containing bitors are Nikkomycins and Polyoxins. Nikkomycin (Fig. 2 16) is
vesicles, and higher activity against both Saccharomyces cerevisiae active against highly chitinous dimorphic fungal pathogens and it
and amphotericin B-resistant Candida albicans strains [44,45]. Over is not active against neither Candida albicans nor C. tropicalis [59].
the last decade, the lipid formulations of amphotericin B have Polyoxins (Fig. 2 17) is active against phytopathogenic fungi [50].
allowed this polyene to be more effective in reducing host toxicity. These are naturally occurring peptidyl nucleoside antifungals that
Formulations include encapsulation in liposomes or in ribbon-like bind to the catalytic site acting as competitive analogs of UDP-N-
and disc-like lipid complexes [46,47]. Other formulations are under acetylglucosamine [6]. However, their current clinical use is com-
investigation, including amphotericin B-cochleate preparations promised due to their hydrolytic liability and low in vivo activity.
[48] and arabinogalactan complexes [49]. Polyoxins are no longer in development since 2012 whereas Nikko-
mycin Z is being tested in clinical trials [9] (Table 1).
2.3. Fungal cell wall synthesis
2.4. Sphingolipids biosynthesis
2.3.1. Inhibitors b-glucan synthesis
Glucans are polysaccharides that consist of D-glucose mono- Sphingolipids are abundant components of membranes in
mers attached to each other by b-(1,3) or b-(1,6)-glucan linkages eukaryotic cells and, besides playing a variety of roles in fungal
[50]. b-(1,3)-D-Glucan constitutes more than 50% of the cell wall cells, some of them play an important role in fungal pathogenesis
and is the main structural polysaccharide to which other cell wall [62]. Recently, some studies have demonstrated that acting on
components (chitins and glycoproteins) are attached. enzymes involved in sphingolipid biosynthesis could attenuate
Currently, the only available antifungal drugs targeting cell wall the virulence of fungal pathogens.
are three echinocandins (caspofungin, micafungin and anidula- Aureobasidin A (Fig. 2 18) is a cyclic depsipeptide isolated from
fungin) [51,52]. Echinocandins are the newest antifungal agents Aureobasidium pullulans [63], active against Saccharomyces cere-
although they were approved for clinical use by FDA and EMA visiae, Schizosaccharomyces pombe, C. albicans, C. glabrata, A. nidu-
(European Medicines Agency) more than a decade ago [7]. They lans, A. niger and C. neoformans, and less effective against A.

Please cite this article in press as: S. Campoy, J.L. Adrio, Antifungals, Biochem. Pharmacol. (2016), http://dx.doi.org/10.1016/j.bcp.2016.11.019
S. Campoy, J.L. Adrio / Biochemical Pharmacology xxx (2016) xxx–xxx 5

Table 1
Antifungal pipeline. Compounds in preclinical or clinical development. QIDP (Qualified Infectious Disease Product).

Mode of action Compound Indication Development References


Ergosterol biosynthesis VT1129 Cryptococcal meningitis Phase 1. Granted QIDP and Orphan www.viamet.com [132]
drug status (US)
VT1161 Onychomycosis, Recurrent vulvovaginal Phase 2b [131]
candidiasis
Isavuconazole Invasive molds disease Candidemia, Phase 3 [129]
invasive candidiasis Phase 3 www.astellas.us
b-Glucan synthesis SCY-078 Invasive candidiasis, vulvovaginal Phase 2. Granted QIDP designation [133]
candidiasis www.scynexis.com
ASP9726 Invasive pulmonary aspergillosis Preclinical [134]
[135]
Biafungin (CD101) Invasive candidiasis, recurrent Phase 2. Granted QIDP and Orphan www.cidara.com
vulvovaginal candidiasis. Drug status (US)
Chitin synthesis Nikkomycin Z Coccidioidomycosis Phase 1. Granted Orphan Drug status www.valleyfeversolutions.com
(US)
Mitocondrial function T-2307 Candidiasis, Cryptococcosis, Phase 1 [136]
aspergillosis www.toyama-chemical.co.jp
Pyrimidine F901318 Invasive aspergillosis Phase 1 [137]
biosynthesis www.f2g.com
GPI anchor APX001 Fusariosis, pulmonary aspergillosis Phase 1 www.amplyx.com
biosynthesis
Carbon metabolism AR-12 (OSU-03012) Cryptococcosis, C. albicans (biofilms). Preclinical. Granted Orphan Drug [139]
status (EU)
Unknown VL2397 Invasive aspergillosis Phase 1 [139]
www.vical.com

fumigatus [64]. Aureobasidin A inhibits the inositol phosphorylce- fungal enzyme for protein synthesis. Tavaborole targets this
ramide (IPC) synthase that transfers the phosphoinositol group enzyme by binding to the editing site together with tRNA so this
from phosphatidylinositol (PI) to the 1-hydroxy group of phytoce- cannot complete the amino acid transfer to the ribosome for
ramide to form IPC [64,65]. The reaction is a key step in fungal sph- assembly and protein synthesis is effectively blocked [72]. Tav-
ingolipid biosynthesis which plays essential roles both as aborole has 1.000-fold higher affinity for the fungal synthetase
structural cell membrane components and cell signaling. IPC syn- than for the human enzyme [71].
thase is not present in mammalian cells, so IPC synthase inhibitors Other b-amino acids to target protein biosynthesis through
could be good candidates to develop antifungal agents [62]. inhibition of isoleucyl tRNA synthetase are Cispentacin [73]
Recently, novel Aureobasidin A derivatives have been synthetized (Fig. 2 21) isolated from the culture broth of a Bacillus cereus strain
by modifying and/or exchanging amino acids in its sequence pro- and its synthetic derivative Icofungipen (Fig. 2 22). Both showed
ducing compounds with high activity against A. fumigatus [66]. high antifungal activity against Candida albicans [74].
Sordarin was isolated from the fermentation broth of the asco-
2.5. Nucleic acid synthesis inhibitors mycete Sordaria araneosa in 1969 [75]. This compound blocks the
function of the fungal, but not human, translation elongation factor
Flucytosine (5-FC; 5-fluorocytosine) is a fluorinated pyrimidine 2 (EF2). The in vitro potency and the spectrum of activity of all sor-
analog with fungistatic activity that interferes with pyrimidine darin analogs depend on the functional group (R) located at the 30 -
metabolism, as well as RNA/DNA and protein synthesis [67]. It is position (Fig. 2 23) [76]. At least, 22 new strains producing a num-
taken up by fungal cells via cytosine permease and converted by ber of different sordarin analogs have been found [77]. Also new
cytosine deaminase to 5-fluorouracil (5-FU) which is transformed semi-synthetic derivatives have been synthetized by replacing
by UMP pyrophosphorylase into 5-fluorouridine monophosphate the glycoside part with different moieties like alkanesulfonate,
(5-FUMP), which is phosphorylated and incorporated into RNA, alkylthio, morpholinyl ring, oxazepane ring or trisubstituted
instead of UTP, resulting in inhibition of protein synthesis. 5-FU tetrahydrofuran ring [78–80]. Its high specificity makes sordarin
(Fig. 2 19) also undergoes conversion into 5-FdUMP (5- very interesting for the development of new derivatives.
fluorodeoxyuridine monophosphate), a potent inhibitor of
thymidylate synthase, that inhibits fungal DNA synthesis and 2.7. Microtubules biosynthesis inhibitors
nuclear division [60,67,68]. This compound is selectively toxic to
fungi as there is little or no cytosine deaminase activity in mam- Microtubules are polymers of a- and b-tubulin dimers that form
malian cells [69]. a highly organized cellular skeleton in all eukaryotic cells [81].
5-FC is active in vitro as well as in vivo against some strains of Antifungal agents like griseofulvin (Fig. 2 24) or vinblastine belong
Candida and Cryptococcus. Most filamentous fungi lack thymidylate this group. Griseofulvin is a natural product isolated from Penicil-
synthase and hence useful spectrum of flucytosine is restricted to lium griseofulvin in 1939 and was the earliest inhibitory agent to
pathogenic yeasts. Resistance is quite commonly seen so 5-FC usu- fungal species [82]. It is toxic to the liver and the spectrum of
ally is used as adjunctive rather than on primary therapy [70]. action is restricted to the dermatophyte fungi (causing ringworm
and athlete’s foot) [83,84]. This compound binds to tubulin, inter-
2.6. Protein biosynthesis inhibitors fering with fungal microtubule assembly and inhibiting mitosis.

Tavaborole (Fig. 2 20) is an oxaborole antifungal approved by 3. Strategies to develop new antifungal compounds
the FDA in 2014 for the topical treatment of toenail onychomycosis
caused by Trichophyton rubrum and T. mentagrophytes. It shows The number of antifungal agents is limited as compared to
antifungal activities against yeast, molds and dermatophytes antibacterial drugs. Fungi are eukaryotic organisms that parasitize
[71]. Tavaborole inhibits the leucyl-tRNA synthetase, an essential eukaryotic hosts and therefore the scarce physiologic differences

Please cite this article in press as: S. Campoy, J.L. Adrio, Antifungals, Biochem. Pharmacol. (2016), http://dx.doi.org/10.1016/j.bcp.2016.11.019
6 S. Campoy, J.L. Adrio / Biochemical Pharmacology xxx (2016) xxx–xxx

between both make more difficult to develop safe and broad spec- Kaltdorf et al. (2016) [93] using bioinformatics tools have iden-
trum antifungal agents. A number of antifungal agents are avail- tified 64 targets in Aspergillus fumigatus that will be evaluated for
able in the market but, excluding amphotericin B and a few more antifungal development. These targets include metabolic enzymes
compounds, almost all them are fungistatic [85]. involved in vitamins, lipids or amino acids biosynthesis that do not
At the present the efforts are focused on identifying new fungal- have close orthologs in humans. Other strategies that are being fol-
specific targets that are critical for fungal growth and have mini- lowed to find new specific fungal targets are those based on tran-
mal similarity to targets among human proteins. scriptome studies in A. fumigatus focused on identify invasion-
Trehalose production seems to be required for the virulence of related gene expression changes [94] and detailed analyses of
several fungal species. Deletion of TPS2 encoding trehalose-6- protein-protein interactions in fungal infections [95].
phosphate phosphatase leads to accumulation of trehalose-6- Historically, the most common approach for identifying anti-
phosphate and cell death. Recently, the crystal structure of C. albi- fungal molecules has been screening large libraries of synthetic
cans Tsp2 has been reported [86] allowing for the identification of small molecules or natural products [1,8,96]. Indeed, two approved
specific inhibitors for this potential target. The signaling molecules antifungal drug classes, the echinocandins and polyenes, were dis-
Ras GTPases play major roles in fungal virulence as they are covered by screening of natural products. Very recently a couple of
required for growth at high temperatures. Therefore, several inhi- examples related to this strategy have been reported. Psoriasin, a
bitors of Ras function, like farnesyltransferase inhibitors, have been small fungicidal protein active against various filamentous fungi
proposed for the specific design of antifungal agents [87]. Cal- including Trichophyton rubrum and A. fumigatus, but not against
cium/calmodulin signaling is another important regulator of stress C. albicans was isolated from lesional psoriasis scale extracts [97].
responses in fungi, including resistance to antifungal treatment. Psoriasin works by chelating free intracellular zinc leading to fun-
Some calcineurin inhibitors are known to have a potent antifungal gal apoptosis. Thus, selective fungal cell-penetrating zinc-chelators
activity although some of them are being used as immunosuppres- could be a new approach to find new antifungal agents.
sant drugs (e.g. tacrolimus). Therefore, development of non- Humidimycin, a ring peptide produced by Streptomyces humidus
immunosuppressive calcineurin inhibitors could be another way that improves the antifungal activity of caspofungin by 10-fold was
to develop antifungal drugs. Another protein being explored as identified from the screening of a collection of 20,000 microbial
antifungal drug target is the Hsp90 heat shock protein since in extracts [98]. Humidimycin acts by inhibiting the fungal high
fungi this protein is involved in azole- and echinocandins- osmolarity glycerol (HOG) response pathway, reducing the protec-
resistance. To date, combination of Hsp90 inhibitors, like gel- tive stress response induced by caspofungin [98]. As the HOG path-
danamycin, with echinocandins or fluconazole improved the fungi- way is not present in human cells, humidimycin is not toxic,
cidal effect [88]. resulting in a promising drug.
Glycosylphosphatidylinositol (GPI) cell wall anchor synthesis Growth-inhibition assays are the most commonly used to iden-
pathway is another promising antifungal target. Novel inhibitors tify antifungal small molecules. However, they have several facts
of Gwt1 and Mcd4, two enzymes in the GPI anchor pathway were that limit their use since many pathogenic fungi grow as filaments
identified by the chemical-genomics-based screening platform making difficult a correlation between growth and OD (optical
CaFT (Candida albicans fitness test) [89]. Screening was performed density). They are not useful for identifying molecules active
against more than 1000 synthetic compounds known to inhibit C. against fungal biofilms and they cannot differentiate molecules
albicans growth but for which their drug target and mechanism that inhibit growth and those that kill the microorganism [8].
of action were unknown. Two compounds, G884 and G365 were Recently, to address such limitations, new high throughput screen-
identified as Gwt1 inhibitors. Both showed activity against C. albi- ing assays have been developed. In many of them, the approach has
cans and only G365 displayed strong activity against A. fumigatus. been to asses loss of cellular integrity using as markers of cell lysis
M743 was identified under screening of natural products extracts. the cytoplasmic enzyme adenylate kinase [99], or dyes such as Ala-
This compound and its semisynthetic derivative M720 are specific marBlue", XTT and resazurine that are converted to fluorescent
inhibitors of Mcd4 and displayed a potent activity against many molecules when they are metabolized by viable cells organisms
Candida and Aspergillus species. [100,101].
The use of CaFT and other genetic platforms, such as the S. cere-
visiae haploinsufficiency profiling (HOP) that offer specific assays
for all possible drug targets in yeast [8] will continue to be extre-
mely helpful in the discovery and mechanism of action of novel 4. Mechanisms of antifungal resistance
antifungals.
Chemical genomics-based approaches have been used to iden- Antifungal resistance is an evolutionary process based on natu-
tify FDA-approved compounds that potentiate the fluconazole ral selection of organisms that enhance their ability to survive and
activity against several yeasts [90]. An antifungal combination grow in the presence of drug. The evolution of resistance against
matrix was developed to analyze around 86,000 chemical interac- antimicrobial agents is ubiquitous in nature and microbes evolve
tions between 3600 small molecules and 6 approved antifungal various strategies to combat the action of drugs.
drugs against four species of fungi [91]. A susceptibility test against The mechanisms of antifungal resistance have been reported at
fluconazole-resistant isolate of C. albicans unveiled combinations the molecular level for most antifungal agents and fungal patho-
capable of potentiating fluconazole activity in this strain. These gens [102–104]. To counteract the fungicidal or fungistatic effects
authors also found that combinations of clofazimine, a compound of all antifungal classes, microorganisms develop three major
with unreported antifungal activity, with several antifungals mechanisms of resistance [7,104]. The first is based on decreasing
exhibited activity against several fungal pathogens. A large gene the effective drug concentration, the second is drug target alter-
deletion library in C. neoformans and a decision-guiding process ations and the third is due to modifications of metabolism to divert
were used to identify and analyze gene-drug interactions that the toxic effects exerted by some antifungal agents.
could help to find drugs that act synergistically against C. neofor- One mechanism to decrease the drug intracellular concentra-
mans [92]. Gene drug interactions were identified for over 80% of tion is to increase drug efflux. Antifungal drug resistance is medi-
the 1500 gene deletion strains tested. Interestingly, data from C. ated by the activity of transport systems like the pleiotropic drug
neoformans were compared with those obtained with S. cerevisiae, resistance (PDR) class of ATP-binding cassette (ABC) transporters,
results revealed very few conserved responses. and transporters of the major facilitator superfamily (MFS).

Please cite this article in press as: S. Campoy, J.L. Adrio, Antifungals, Biochem. Pharmacol. (2016), http://dx.doi.org/10.1016/j.bcp.2016.11.019
S. Campoy, J.L. Adrio / Biochemical Pharmacology xxx (2016) xxx–xxx 7

CDR1 and CDR2 are the major transporters involved in azole tion. Alteration of the last steps of the ergosterol biosynthesis
resistance in C. albicans. Upregulation enhances drug efflux and through inactivation of the ERG3 gene results in no production of
reduces their accumulation in the cells [105,106]. Many others toxic methylated sterols that lead to cross resistance to all azole
ABC transporters involved in azole resistance have been described drugs [104]. Furthermore, mutations in non-essential genes of this
in C. glabrata (CgCDR1, CgCDR2, CgSNQ2) and AFR1 in C. neofor- pathway (ERG3, ERG6, ERG24 and ERG2) also lead to a decrease, or
mans. In A. fumigatus and A. nidulans ABC transporters have been even total absence, of ergosterol in plasma membrane [128]. As
identified as a resistance mechanism to azole. MDR1 is the MFS reported above mutations in the FUR1 gene decrease the conver-
involved in the development of azole resistance in clinical isolates sion of 5FU into toxic metabolites able to enter the cytosine meta-
form C. albicans and C. dubliniensis and its upregulation results in bolism and exert their toxic effect. Downregulation of FUR1 also
increased azole efflux [107]. FLU1 from C. albicans is another MFS decreased 5FC susceptibility. A 4-fold lower expression of this gene
involved in fluconazole resistance [108]. leads to total resistance to 5FC in C. glabrata species [7].
Upregulation of ABC and MFS transporters is mediated by speci- In the recent years reports on novel resistance profiles have
fic regulations in resistant fungal pathogens. Point mutations been reported and the most problematic is the development of
defined as gain-of-function mutations (GOF) in these regulators simultaneous resistance to at least two different classes of antifun-
confer an inherent high expression level of the transporters in gal agents (a condition known as multidrug resistance, MDR) [104].
the drug-resistance strains [7]. GOF mutations in the transcription Loss of function point mutations or simultaneous mutations in
factor Upc2p [109] caused increased fluconazole resistance in C. ERG2, ERG3, ERG5 and ERG11 result in MDR to azoles and ampho-
albicans. In the same way, upregulation of Cyp51A in A. fumigatus tericin B in Candida species [104,128].
lead to azole resistance mediated by duplication of 34 and 42 bp Resistance to echinocandins (e.g. caspofungin) and azoles has
in the Cyp51A promoter [110]. also been reported. The resistance mechanisms seem to be due
Another way to decrease the effective drug concentration is to combinations of mutations in the FSK2 gene and ABC trans-
overexpression of the ERG11 (gene encoding 14-a sterol demethy- porters upregulation [104]. Although not very common, a few
lase) and/or other genes of sterol biosynthesis, such as ERG1, ERG3, cases of resistance for more than two drug classes have been
ERG7, ERG9 or ERG25 which encode enzymes downstream C14a- recently described in C. albicans and C. lusitaniae isolates [144].
demethylase such upregulation leads to azole resistance The current trends show that the highest proportion of resistant
[99,111,112,113]. isolates is from C. glabrata [9] representing more than 10% of total
Another mechanism to reduce the effective drug concentration isolates in the European Union and more than 20% in the USA. Such
in some medically relevant fungi including Candida, Aspergillus, isolates show high resistance to azoles (fluconazole and voricona-
Cryptococcus, Trichosporon, Coccidioides and Pneumocystis zole) as well as to echinocandins. New infections due to Fusarium
[114,115] is the ability to form biofilms increasing their resistance spp., Zygomycetes and azole- and echinocandins-resistant C. glab-
to several drugs including azoles, pyrimidine analogs and polyenes rata isolates are being more frequently observed.
[116]. Biofilms are multicellular structures in which cells form a
dense network that is covered by a matrix of polysaccharides, car-
bohydrates, proteins and signaling molecules that restricts the 5. Antifungal pipeline
penetration of drugs by formation of a diffusion barrier sequester-
ing the antifungal agents [117–120]. Despite it is clear that there is an urgent need for new antifun-
Only amphotericin B and the echinocandins (e.g. caspofungin gal agents [8–10,96,104] in their complex development as well as
and micafungin) have demonstrated consistent in vitro activity the lack of investment are responsible for a limited drug develop-
against C. albicans biofilms [121]. However, even with these two ment pipeline. However, antifungal development has been recently
agents, Candida biofilm-related infections are extremely difficult, boosted by two Acts from the US Administration. The first is GAIN
if not impossible, to eradicate [122]. (Generating Antibiotic Incentives Now) that provides a fast track
Target drug alteration is another mechanism by which fungal description and priority review by FDA to QIDP (Qualified Infec-
pathogens are able to develop resistance. This mechanism has been tions Disease Products) as well as a 5 year extension of market
reported for azoles and echinocandins. Mutations in ERG11 in C. exclusivity. The second is the Orphan Drug Act that encourages
albicans decrease affinity for azoles leading to resistance. Many of the development of drugs for rare diseases, including several inva-
these point mutations decreased affinity to fluconazole and have sive mycoses, and provides 7 year market exclusivity.
a moderate effect on posaconazole [123]. In A. fumigatus single The new antifungal drugs should improve fungal disease mor-
mutations in Cyp51A are the most frequent mechanism to confer tality, show better fungicidal activity, extend the spectrum of
high level resistance to azoles in clinical isolates of this species activity against drug-resistant fungi and rare molds such as Sce-
[124]. dosporium spp., improve the pharmacokinetics and pharmacody-
Echinocandin resistance is systematically associated with point namics, reduce/avoid host toxicity, cause few drug-drug
mutations in either FSK1 or FSK2 genes [7,104]. These mutations interactions, and act by new, selective and different mechanisms
are located in two different not spot regions of these genes named of action. Indeed, no new classes of antifungals have been
HS1 and HS2. Hot spot mutations have been reported in C. albicans, approved since 2006 when the FDA and the EMA (European
C. glabrata, C. tropicalis, C. krusei, Scedosporium apiospermum and A. Medicines Agency) authorized anidulafungin.
fumigatus [7,104]. Such mutations alter the kinetics of the glucan The current antifungal pipeline contains several categories of
synthase resulting in significantly higher inhibitory concentrations compounds at different stages of development [1,9,10,96]. In
and inhibition constant [125,126]. Table 1 we summarize the most promising antifungal molecules
The most common mechanism of resistance to pyrimidine ana- that are in preclinical and in clinical development.
logs (5FC) is a point mutation in the FUR1 gene. This mutation leads Several compounds in the pipeline are derivatives of the azoles,
to complete resistance to 5FC and 5FU in fungi [7]. A second point echinocandins and polyenes and, therefore, they have the same
mutation in the gene encoding for cytosine deaminase results in mechanism of action targeting ergosterol biosynthesis, membrane
5FC resistance in many Candida strains [127]. disruption and cell wall synthesis (b-1,3-D-glucan synthesis),
The third mechanism by which fungal pathogens can develop respectively.
resistance against antifungal agents is by modifying metabolic Isavuconazole, the most recently approved triazole, is being
pathways leading to loss or strong decrease of their specific func- evaluated in two different phase 3 trials. The results from the first

Please cite this article in press as: S. Campoy, J.L. Adrio, Antifungals, Biochem. Pharmacol. (2016), http://dx.doi.org/10.1016/j.bcp.2016.11.019
8 S. Campoy, J.L. Adrio / Biochemical Pharmacology xxx (2016) xxx–xxx

trial to assess the efficacy and safety of isavuconazole versus conserved in humans, human URA1 is only 20% identical to its fun-
voriconazole for the treatment of invasive molds diseases caused gal homolog and it is inhibited 2000-fold less effectively by this
by Aspergillus and other filamentous fungi (SECURE) have been drug. F901318 is highly active against azole and amphotericin B-
recently reported. Results showed that isavuconazole was not infe- resistant Aspergillus strains but is inactive against the Mucorales
rior to voriconazole for the primary treatment of such disease and and Candida spp. F901318 is currently in phase 1 clinical trials
it was well tolerated compared with voriconazole, with fewer for invasive aspergillosis.
study drug-related adverse effects [129]. Isavuconazole is also cur- APX001A (previously named E1210) is an inhibitor of the fungal
rently in a second phase 3 clinical trial to evaluate its efficacy and Gwt1 protein that catalyzes an early step in the biosynthesis of the
safety of intravenous and oral formulations for the treatment of glycosylphosphatidylinositol (GPI) anchor that is required for the
candidemia and invasive Candida infections (ACTIVE) [www.astel- anchoring of mannosylated proteins of the fungal cell wall and cell
las.us]. membrane. It has activity against Candida, Aspergillus, Fusarium and
VT1129 and VT1161 belong to a novel class of triazoles that Scedosporium in vitro. It also shows activity against azole and
were identified as part of an effort trying to find molecules with echinocandin-resistant strains of Candida [143]. Very recently,
very low affinity to human CYP enzymes, including human APX001 has been granted QIDP designation by FDA. This product
CYP51. In vitro, VT1161 is about 1000-fold more selective for Can- is currently in phase 1 clinical trials for the treatment of invasive
dida CYP51 than for the human enzyme, while VT1129 is approxi- aspergillosis, invasive candidiasis, and coccidioidomycosis.
mately 3000-fold more selective for the Cryptococcus isoforms over AR-12 (OSU-03012) was identified through a repurposing
human [130]. VT1129 inhibits the growth of Cryptococcus neofor- screen of protein kinase inhibitors looking for antifungal agents.
mans and C. gattii, and in a mouse model of cryptococcal meningitis It has been granted the European Orphan Drug status for the treat-
performed better than fluconazole. This molecule has been granted ment of cryptococcosis. AR-12 inhibits acetyl-CoA synthetase that
QIDP designation and orphan drug status by FDA and is in phase 1 leads to defects in several cellular processes [138]. It is in preclin-
clinical trials for the treatment of cryptococcal meningitis. VT1161 ical development for the treatment of cryptococcosis.
which is very effective against fluconazole-resistant Candida iso- VL-2397 is a new class of antifungal that has received QIDP des-
lates is in phase 2b trials for the treatment of onychomycosis and ignation for development for treatment of aspergillosis. Its mecha-
recurrent vulvovaginal candidiasis [131]. nism of fungal cell inhibition is not known, although it seems that
SCY-078 is a novel b-1,3-glucan synthase inhibitor structurally the import of this drug into fungal cells requires the siderophore
different than the current echinocandins. It is a derivative from transporter Sit1. As this transporter is absent in mammalian cells,
enfumafungin, a triterpene glycoside produced by fermentation this mechanism of entry could be specific for fungal pathogens. VL-
of a Hormonema sp. [140]. SCY-078 shows activity against 2397 shows fungicidal activity against Aspergillus, but it has no
echinocandin-resistant isolates of Candida and Aspergillus [141], activity against Candida albicans or other Candida species. It is cur-
against non-Aspergillus molds comparable to that of echinocandins rently in phase 1 clinical trials for the treatment of invasive
and also against Scedosporium prolificans, a resistant mold for aspergillosis.
which there are no good treatment options. It is also interesting
the good activity against Paecilomyces variotii, fungi associated
with disseminated infections (fungemia and peritonitis in patients
undergoing peritoneal dialysis) [133]. It has received QIDP desig- 6. Conclusions
nation and its oral formulation is currently in phase 2 clinical trials
for the treatment of invasive candidiasis, while its IV (intra- Invasive fungal infections have remarkably increased in the last
venously) formulation is in phase 1. Novel semi-synthetic enfuma- few years and the number of patients at risk for these infections is
fungin derivatives are been studied to improve antifungal potency increasing as immunomodulatory therapies continue to expand.
and oral pharmacokinetic properties [142]. Despite the numerous available antifungal drugs, they do not
Biafungin (CD101) is a novel echinocandin that shows similar meet the expectations for managing these fungal infections. Mor-
activity to caspofungin and anidulafungin against Aspergillus and tality rates are still unacceptably high, increasing azole- and
Candida species, but its advantage lies in its pharmacokinetics. Its echinocandin-resistant strains are being reported more frequently,
half-time life is about 4-fold longer (81 h) than the half-time of and acute and chronic side effects are often observed. Therefore, as
anidulafungin (24 h), the longest-acting echinocandin to date. Like reported and agreed by many researchers there is an emerging
other echinocandins, it shows few drug interactions and excellent need to fill the pipeline with new antifungal drugs [8–10,96].
safety profile. Biafungin has been granted QIDP designation and However, although antifungal development has been pushed
orphan drug status and it is currently in phase 2 clinical trials for forward by FDA, through GAIN (QIDP designation) and Orphan
the treatment of candidemia. Drug Acts, the pipeline only shows a few compounds with novel
Nikkomycin Z is a competitive inhibitor of chitin synthesis. It modes of action that are in preclinical development or in early clin-
has been recently received Orphan Drug status and it is being ical trials. Therefore, it is quite hard to predict which (if any) of
tested in phase 1 clinical trials for treatment for these molecules will emerge as a new clinical antifungal. Due to
coccidioidomycosis. the challenges that the development of broad spectrum antifungals
Very interestingly, there are some new candidates that show take, the development of narrow-spectrum drugs could make the
either different modes of action from the currently marketed early stages of drug development more straightforward. Recent
drugs, or that even have not been identified yet. T-2307 is a novel gene–drug interaction dataset analysis suggest that focusing on
arylamidine that collapses fungal mitochondrial membrane com- antifungal development that is geared toward particular patho-
promising energy production for essential cellular processes gens, may lead to more potent and effective therapies for invasive
[136]. Interestingly, this activity is specific to fungi as it seems to fungal infections.
be due to the selective uptake of this drug by the high-affinity fun- The identification of new therapeutic targets by use of genomic
gal specific Agp2 spermine-spermidine transporter. T-2307 under- techniques, and the expanding knowledge on the cellular processes
went successful phase 1 trials for the treatment of candidiasis. required for fungal survival and virulence will help to find and
F901318 is an orotomide, a novel class of antifungals, which develop compounds with novel mode of actions. The repurposing
inhibits pyridine biosynthesis by blocking dihydroorotate of available drugs for their use as antifungal agents and new
dehydrogenase (URA1) activity [137]. Although this pathway is derivatives from azoles, echinocandins and polyenes, as well as

Please cite this article in press as: S. Campoy, J.L. Adrio, Antifungals, Biochem. Pharmacol. (2016), http://dx.doi.org/10.1016/j.bcp.2016.11.019
S. Campoy, J.L. Adrio / Biochemical Pharmacology xxx (2016) xxx–xxx 9

screening for new natural products, will help to expand the cur- [32] M.A. Hossain, M.A. Ghannoum, New developments in chemotherapy for non-
invasive fungal infections, Expert Opin. Investig. Drugs 10 (2001) 1501–1511.
rently limited and available antifungal pipeline.
[33] D. Andes, In vivo pharmacodynamics of antifungal drugs in treatment of
candidiasis, Antimicrob. Agents Chemother. 47 (2003) 1179–1186.
References [34] T.M. Anderson, M.C. Clay, A.G. Cioffi, K.A. Diaz, G.S. Hisao, M.D. Tuttle, et al.,
Amphotericin forms an extramembranous and fungicidal sterol sponge, Nat.
Chem. Biol. 10 (2014) 400–406, http://dx.doi.org/10.1038/nchembio.1496.
[1] K.M. Pianalto, J.A. Alspaugh, New horizons in antifungal therapy, J. Fungi 2
[35] Z. Sklenár, V. Scigel, K. Horáckova, O. Slanar, Compounded preparations with
(2016) 1–24.
nystatin for oral and oromucosal administration, Acta Pol. Pharm. Drug Res.
[2] G.D. Brown, D.W. Denning, N.A.R. Gow, S.M. Levitz, M.G. Netea, T.C. White,
70 (2013) 759–762.
Hidden killers: human fungal infections, in: Sci. Transl. Med. 4 (2012)
[36] P. Lalitha, V.R. Kumar, N.V. Prajna, A.W. Fothergill, In vitro natamycin
165rv13.
susceptibility of ocular isolates of Fusarium and Aspergillus species:
[3] D. Diekema, S. Arbefeville, L. Boyken, J. Kroeger, M. Pfaller, The changing
comparison of commercially formulated natamycin eye drops to
epidemiology of healthcare-associated candidemia over three decades, Diagn.
pharmaceutical-grade powder, J. Clin. Microbiol. 46 (10) (2008) 3477–3478.
Microbiol. Infect. Dis. 73 (2012) 45–48.
[37] M.A. Farid, H.A. El-Enshasy, A.I. El-Diwany, E.S.A. El-Sayed, Optimization of
[4] K.A. Marr, H.T. Schlamm, R. Herbrecht, S.T. Rottinghauscott, E.J. Bow, O.A.
the cultivation medium for natamycin production by Streptomyces natalensis,
Cornely, et al., Combination antifungal therapy for invasive aspergillosis, Ann.
J. Basic Microbiol. 40 (2000) 157–166.
Intern. Med. 162 (2015) 81–89.
[38] S.B. Zotchev, Polyene macrolide antibiotics and their applications in human
[5] D.W. Denning, W.W. Hope, Therapy for fungal diseases: opportunities and
therapy, Curr. Med. Chem. 10 (2003) 211–223.
priorities, Trends Microbiol. 18 (2010) 195–204.
[39] R. Laniado-Laborín, M.N. Cabrales-Vargas, Amphotericin B: side effects and
[6] M.V. Castelli, E. Butassi, M.C. Monteiro, L.A. Svetaz, F. Vicente, A. Susana, S.A.
toxicity, Rev. Iberoam. Micol. 26 (2009) 223–227.
Zacchino, Novel antifungal agents: a patent review (2011-present), Expert
[40] P. Caffrey, S. Lynch, E. Flood, S. Finnan, M. Oliynyk, Amphotericin biosynthesis
Opin. Ther. Pat. 24 (2014) 1–16.
in Streptomyces nodosus: deductions from analysis of polyketide synthase and
[7] P. Vandeputte, S. Ferrari, A.T. Coste, Antifungal resistance and new strategies
late genes, Chem. Biol. 8 (2001) 713–723.
to control fungal infections, Int. J. Microbiol. (2012) 26, http://dx.doi.org/
[41] A. Lemke, A.F. Kiderlen, O. Kayser, Amphotericin B, Appl. Microbiol.
10.1155/2012/713687. ID 713687.
Biotechnol. 68 (2005) 151–162.
[8] T. Roemer, D.J. Krysan, Antifungal drug development: challenges, unmet
[42] F.C. Odds, A.J.P. Brown, N.A.R. Gow, Antifungal agents: mechanisms of action,
clinical needs, and new approaches, Cold Spring Harb. Perspect. Med. 4 (2014)
Trends Microbiol. 11 (2003) 272–279.
a019703.
[43] W.J. Gibbs, R.H. Drew, J.R. Perfect, Liposomal amphotericin B: clinical
[9] D.W. Denning, M.J. Bromley, How to bolster the antifungal pipeline, Science
experience and perspectives, Expert Rev. Anti Infect. Ther. 3 (2005) 167–181.
347 (2015) 1414–1416.
[44] R.D. Santo, Natural products as antifungal agents against clinically relevant
[10] J.R. Perfect, Is there an emerging need for new antifungals? Expert Opin.
pathogens, Nat. Prod. Rep. 27 (2010) 1084–1098.
Emerg. Drugs 21 (2016) 129–131, http://dx.doi.org/10.1517/
[45] H. Kakeya, Y. Miyazaki, H. Senda, T. Kobayashi, M. Seki, K. Izumikawa, et al.,
14728214.2016.1155554.
Efficacy of SPK-843, a novel polyene antifungal, in comparison with
[11] D.J. Sheehan, C.A. Hitchcock, C.M. Sibley, Current and emerging azole
amphotericin B, liposomal amphotericin B, and micafungin against murine
antifungal agents, Clin. Microbiol. Rev. 12 (1999) 40–79.
pulmonary aspergillosis, Antimicrob. Agents Chemother. 52 (2008) 1868–
[12] M.A. Ghannoum, Future of antimycotic therapy, Dermatol. Therapy 3 (1997)
1870.
104–111.
[46] B. Dupont, Overview of the lipid formulations of amphotericin B, J.
[13] A.J. Carrillo-Munoz, S. Giusiano, P.A. Ezkurra, G. Quindos, Antifungal agents:
Antimicrob. Chemother. 49 (2002) 31–36.
mode of action in yeast cells, Rev. Esp. Quimioter. 19 (2006) 130–139.
[47] P. Chandrasekar, Management of invasive fungal infections: a role for
[14] J.H. Dawson, M. Sono, Cytochrome P-450 and chloroperoxidase: thiolate-
polyenes, J. Antimicrob. Chemother. 66 (2011) 457–465.
ligated heme enzymes. Spectroscopic determination of their active-site
[48] L. Zarif, J.R. Graybill, D. Perlin, R.J. Mannino, Cochleates: new lipid-based drug
structures and mechanistic implications of thiolate ligation, Chem. Rev. 87
delivery system, J. Liposome Res. 10 (2000) 523–538.
(1987) 1255–1276.
[49] R. Falk, A.J. Domb, I. Polacheck, A novel injectable water-soluble amphotericin
[15] H.V. Bossche, L. Koymans, H. Moereels, P450 inhibitors of use in medical
B-arabinogalactan conjugate, Antimicrob. Agents Chemother. 43 (1999)
treatment: focus on mechanisms of action, Pharmacol. Ther. 67 (1995) 79–
1975–1981.
100.
[50] T. Lorand, B. Kocsis, Recent advances in antifungal agents, Mini Rev. Med.
[16] P.K. Shukla, P. Singh, R.K. Yadav, S. Pandey, S.S. Bhunia, Past, present and
Chem. 7 (2007) 900–911.
future of antifungal drug development, Top. Med. Chem. (2016), http://dx.doi.
[51] A.J. Sucher, E.B. Chahine, H.E. Balcer, Echinocandins: the newest class of
org/10.1007/7355_2016_4.
antifungals, Ann. Pharmacother. 43 (2009) 1647–1657.
[17] M.T. Nozawa, Effects of antifungal agents on ergosterol biosynthesis in
[52] P.K. Mukherjee, D. Sheehan, L. Puzniak, H. Schlamm, M.A. Ghannoum,
Candida albicans and Trichophyton mentagrophytes: differential inhibitory
Echinocandins: are they all the same? J. Chemother. 23 (2011) 319–325,
sites of naphthiomate and miconazole, J. Invest. Dermatol. 85 (1985) 434–
http://dx.doi.org/10.1179/joc.2011.23.6.319.
437.
[53] J.A. Vazquez, J.D. Sobel, Anidulafungin: a novel echinocandin, Clin. Infect. Dis.
[18] A. Pardasani, Oral antifungal agents used in dermatology, Curr. Probl.
43 (2006) 215–222.
Dermatol. 11 (2000) 270.
[54] G. Eschenauer, D.D. DePestel, P.L. Carver, Comparison of echinocandin
[19] P.G. Pappas, J.H. Rex, J.D. Sobel, S.G. Filler, W.E. Dismukes, T.J. Walsh, J.E.
antifungals, Ther. Clin. Risk Manag. 3 (2007) 71–97.
Edwards, Guidelines for treatment of candidiasis, Clin. Infect. Dis. 38 (2004)
[55] J.M. Valgus, What’s new in antifungals? Curr. Infect. Dis. Rep. 5 (2003) 16–21.
161–189.
[56] D. Andes, K. Marchillo, J. Lowther, A. Bryskier, T. Stamstad, R. Conklin, In vivo
[20] T.A. Vincent, Current and future antifungal therapy: new targets for
pharmacodynamics of HMR 3270, a Glucan synthase inhibitor, in a
antifungal therapy, Int. J. Antimicrob. Agents 16 (2000) 317.
murine candidiasis model, Antimicrob. Agents Chemother. 47 (2003)
[21] R. Zaragoza, J. Pemán, The diagnostic and therapeutic approach to fungal
1187–1192.
infections in critical care settings, Adv. Sepsis 6 (2008) 90.
[57] M.A. Pfaller, S.A. Messer, P.R. Rhomberg, R.N. Jones, M. Castanheira, Activity of
[22] G. Petrikkos, A. Skiada, Recent advances in antifungal chemotherapy, Int. J.
a long-acting echinocandin, CD101, determined using CLSI and EUCAST
Antimicrob. Agents 30 (2007) 108–117.
reference methods, against Candida and Aspergillus spp., including
[23] R.T. George, C. Jose, F.P. Thomas, Overview of anti-fungal agents, Clin. Chest
echinocandin- and azole-resistant isolates, J. Antimicrob. Chemother.
Med. 30 (2009) 203.
(2016), http://dx.doi.org/10.1093/jac/dkw214.
[24] V. Nagappan, S. Deresinski, Posaconazole: a broad-spectrum triazole
[58] P. Gershkovich, E. Wasan, M. Lin, O. Sivak, C.G. Leon, J.G. Clement, K.M.
antifungal agent, Clin. Infect. Dis. 45 (2007) 1610–1617.
Wasan, Pharmacokinetics and biodistribution of amphotericin B in rats
[25] L.R. Peyton, S. Gallagher, M. Hashemzadeh, Triazole antifungals: a review,
following oral administration in a novel lipid-based formulation, J.
Drugs Today 51 (2015) 705–718.
Antimicrob. Chemother. 64 (2009) 101–108, http://dx.doi.org/10.1093/jac/
[26] M.S. Abdel-Kader, M.M. Muharram, New microbial Source of the antifungal
dkp140.
allylamine ‘‘Terbinafine”, Saudi Pharm. J. (2016), http://dx.doi.org/10.1016/j.
[59] R.A. Akins, An update on antifungal targets and mechanisms of resistance in
jsps.2016.06.006.
Candida albicans, Med. Mycol. 43 (2005) 285–318.
[27] A.K. Gupta, J.E. Ryder, E.A. Cooper, Naftifine: a review, J. Cutan. Med. Surg. 12
[60] M.P. Arevalo, A.J. Carillo-Munoz, J. Salgado, D. Cardenes, S. Brió, G. Quindós, A.
(2008) 51–58.
Espinel-Ingroff, Antifungal activity of the echinocandin anidulafungin
[28] M. Ghannoum, N. Isham, A. Verma, S. Plaum, A. Fleischer Jr., B. Hardas, In vitro
(VER002, LY-303366) against yeast pathogens: a comparative study with
antifungal activity of naftifine hydrochloride against dermatophytes,
M27-A microdilution method, J. Antimicrob. Chemother. 51 (2003) 163–166.
Antimicrob. Agents Chemother. 57 (2013) 4369–4372.
[61] M.F. Vicente, A. Basilio, A. Cabello, F. Peláez, Microbial natural products as a
[29] D.G. Sant, S.G. Tupe, C.V. Ramana, M.V. Deshpande, Fungal cell membrane-
source of antifungals, Clin. Microbiol. Infect. 9 (2003) 15–32.
promising drug target for antifungal therapy, J. Appl. Microbiol. (2016),
[62] R. Rollin-Pinheiro, A. Singh, E. Barreto-Bergter, M. Del Poeta, Sphingolipids as
http://dx.doi.org/10.1111/jam.13301. ISSN 1364-5072.
targets for treatment of fungal infections, Future Med. Chem. 8 (2016) 1469–
[30] D.L. Mayers, Antimicrobial Drug Resistance: Mechanism of drug Resistance,
1484.
vol. 1, Humana Press/Springer, Totowa/New York, 2009, p. 299.
[63] K. Takesako, K. Ikai, F. Haruna, M. Endo, K. Shimanaka, E. Sono, et al.,
[31] M.A. Hossain, M.A. Ghannoum, New investigational antifungal agents for
Aureobasidins, new antifungal antibiotics taxonomy, fermentation, isolation
treating invasive fungal infections, Expert Opin. Investig. Drugs 9 (2000)
and properties, J. Antibiot. 44 (1991) 919–924.
1797–1813.

Please cite this article in press as: S. Campoy, J.L. Adrio, Antifungals, Biochem. Pharmacol. (2016), http://dx.doi.org/10.1016/j.bcp.2016.11.019
10 S. Campoy, J.L. Adrio / Biochemical Pharmacology xxx (2016) xxx–xxx

[64] W. Zhong, M. Jeffries, N.H. Georgopapadakou, Inhibition of inositol [91] N. Robbins, M. Spitzer, T. Yu, R.P. Cerone, A.K. Averette, Y.S. Bahn, et al., An
phosphorylceramide synthase by aureobasidin A in Candida and Aspergillus antifungal combination matrix identifies a rich pool of adjuvant molecules
species, Antimicrob. Agents Chemother. 44 (2000) 651–653. that enhance drug activity against diverse fungal pathogens, Cell Rep. 13
[65] T. Hashida-Okado, A. Ogawa, M. Endo, R. Yasumoto, K. Takesako, I. Kato, AUR1, (2015) 1481–1492.
a novel gene conferring aureobasidin resistance on Saccharomyces cerevisiae: [92] J.C.S. Brown, J. Nelson, B. Vandersluis, R. Deshpande, A. Butts, S. Kagan, et al.,
a study of defective morphologies in Aur1p-depleted cells, Mol. Gen. Genet. Unraveling the biology of a fungal meningitis pathogen using chemical
(Tokyo) 251 (1996) 236–244. genetics, Cell 159 (2014) 1168–1187.
[66] P.G.M. Wuts, L.J. Simons, B.P. Metzger, R.C. Sterling, J.L. Slightom, A.P. [93] M. Kaltdorf, M. Srivastava, S.K. Gupta, C. Liang, J. Binder, A. Dietl, et al.,
Elhammer, Generation of broad-spectrum antifungal drug candidates from Systematic identification of anti-fungal drug targets by a metabolic network
the natural product compound aureobasidin A, ACS Med. Chem. Lett. 6 (2015) approach, Front. Mol. Biosci. 3 (2016) 22, http://dx.doi.org/10.3389/
645–649. fmolb.2016.00022.
[67] J. Onishi, M. Meinz, J. Thompson, J. Curotto, S. Dreikorn, M. Rosenbach, et al., [94] M. Schrettl, N. Beckmann, J. Varga, T. Heinekamp, I.D. Jacobsen, C. Jöchl, et al.,
Discovery of novel antifungal (1,3)-b-D-Glucan synthase inhibitors, HapX-mediated adaption to iron starvation is crucial for virulence of
Antimicrob. Agents Chemother. 44 (2000) 368–377. Aspergillus fumigatus, PLoS Pathol. 6 (2010) e1001124, http://dx.doi.org/
[68] J.A. Dowell, W. Knebel, T. Ludden, M. Stogniew, D. Krause, T. Henkel, 10.1371/journal.ppat.1001124.
Population pharmacokinetic analysis of anidulafungin, an echinocandin [95] F. Lamoth, P.R. Juvvadi, E.J. Soderblom, M.A. Moseley, W.J. Steinbach, Hsp70
antifungal, in: Clin. Pharmacol. 44 (2004) 590–598. and the cochaperone StiA (Hop) orchestrate Hsp90-mediated caspofungin
[69] C. Zhao, T. Huang, W. Chen, Z. Deng, Enhancement of the diversity of tolerance in Aspergillus fumigatus, Antimicrob. Agents Chemother. 59 (2015)
polyoxins by a thymine-7-hydroxylase homolog outside the polyoxin 4727–4733, http://dx.doi.org/10.1128/AAC.00946-15.
biosynthesis gene cluster, Appl. Environ. Microbiol. 76 (2010) 7343–7347. [96] N. Osherov, D.P. Kontoyiannis, The anti-Aspergillus drug pipeline: is the glass
[70] G.G. Zhanel, J.A. Karlowsky, G.A. Harding, T.V. Balko, S.A. Zelenitsky, M. half full or empty? Med. Mycol. (2016), http://dx.doi.org/10.1093/mmy/
Friesen, A. Kabani, M. Turik, D.J. Hoban, In vitro activity of a new myw060.
semisynthetic echinocandin, LY-303366, against systemic isolates of [97] K.Z. Heina, H. Takahashia, T. Tsumorib, Y. Yasuic, Y. Nanjohd, T. Togad, et al.,
Candida species, Cryptococcus neoformans, Blastomyces dermatitidis, and Disulphide-reduced psoriasin is a human apoptosis inducing broad-spectrum
Aspergillus species, Antimicrob. Agents Chemother. 41 (1997) 863–865. fungicide, Proc. Natl. Acad. Sci. U.S.A. 112 (2015) 13039–13044.
[71] A.K. Gupta, S.G. Versteeg, Tavaborole – a treatment for onychomycosis of the [98] V. Valiante, M. Cândida Monteiro, J. Martín, R. Altwasser, N. El Aouad, I.
toenails, Expert Rev. Clin. Pharmacol. 9 (2016) 1145–1152. González, et al., Hitting the caspofungin salvage pathway of human-
[72] F.L. Rock, W. Mao, A. Yaremchuk, M. Tukalo, et al., An antifungal agent pathogenic fungi with the novel lasso peptide humidimycin (MDN-0010),
inhibits an aminoacyl-tRNA synthetase by trapping tRNA in the editing site, Antimicrob. Agents Chemother. 59 (2015) 5145–5153.
Science 316 (2007) 1759–1761. [99] L. Romani, Immunity to fungal infections, Nat. Rev. Immunol. 4 (2004) 1–23.
[73] M. Konishi, M. Nishio, K. Saitoh, T. Miyaki, T. Oki, H. Kawaguchi, Cispentacin, a [100] J.L.A. Rabjohns, Y.D. Park, J. Dehdashti, C. Henderson, A. Zelazny, S.J. Metallo,
new antifungal antibiotic, J. Antibiot. 42 (1989) 1749–1755. et al., A high throughput screening assay for fungicidal compounds against:
[74] V. Petraitis, R. Petraitiene, A.M. Kelaher, A.A. Sarafandi, T. Sein, D. Mickiene, J. Cryptococcus neoformans, J. Biomol. Screen. 19 (2014) 270–277.
Bacher, A.H. Groll, T.J. Walsh, Efficacy of PLD-118, a novel inhibitor of Candida [101] M.C. Monteiro, M. de la Cruz, J. Cantizani, C. Moreno, J.R. Tormo, E. Mellado,
isoleucyl-tRNA synthetase, against experimental oropharyngeal and et al., A new approach to drug discovery: high-throughput screening of
esophageal candidiasis caused by fluconazole-resistant C. albicans, microbial natural extracts against Aspergillus fumigatus Using Resazurin, J.
Antimicrob. Agents Chemother. 48 (2004) 3959–3967. Biomol. Screen. 17 (2012) 542–549.
[75] H. Liang, Sordarin, an antifungal agent with a unique mode of action, Beilstein [102] Z.A. Kanafani, J.R. Perfect, Antimicrobial resistance: resistance to antifungal
J. Org. Chem. 4 (2008) 1–14. agents: mechanisms and clinical impact, Clin. Infect. Dis. 46 (2008) 120–128.
[76] E. Herreros, C.M. Martinez, M.J. Almela, M.S. Marriott, F. Gomez de las Heras, [103] J. Peman, E. Canton, A. Espinel-Ingroff, Antifungal drug resistance
D. Gargallo-Viola, Sordarins: in vitro activities of new antifungal derivatives mechanisms, Expert Rev. Anti Infect. Ther. 7 (2009) 453–460.
against pathogenic yeasts, Pneumocystis carinii and filamentous fungi, [104] D. Sanglard, Emerging threats in antifungal-resistant fungal pathogens, Front.
Antimicrob. Agents Chemother. 42 (1998) 2863–2869. Med. 3 (2016) 11, http://dx.doi.org/10.3389/fmed.2016.00011.
[77] F. Vicente, A. Basilio, G. Platas, J. Collado, G.F. Bills, A. González del Val, et al., [105] N.N. Mishra, T. Prasad, N. Sharma, A. Payasi, R. Prasad, D.K. Gupta, R. Singh,
Distribution of the antifungal agents sordarins across filamentous fungi, Pathogenicity and drug resistance in Candida albicans and other yeast species.
Mycol. Res. 113 (2009) 754–770. A review, Acta Microbiol. Immunol. Hung. 54 (2007) 201–235.
[78] M. Serrano-Wu, X. Du, N. Balasubramanian, D.R. St. Laurent, Thio derivatives [106] J.C. Sardi, A.M. Almeida, M.J. Mendes Giannini, New antimicrobial therapies
of sordarin as antifungal agents, US 6,436,930 B1 (2002). used against fungi present in subgingival sites – a brief review, Arch. Oral
[79] S. Kaneko, M. Arai, T. Uchida, T. Harasaki, T. Fukuoka, T. Konosu, Synthesis and Biol. 56 (2011) 951–959.
evaluation of N-substituted 1,4-oxazepanyl sordaricins as selective fungal EF- [107] S. Mogavero, A. Tavanti, S. Senesi, P.D. Rogers, J. Morschhäuser, Differential
2 inhibitors, Bioorg. Med. Chem. Lett. 12 (2002) 1705–1708. requirement of the transcription factor Mcm1 for activation of the Candida
[80] T. Hanadate, M. Tomishima, N. Shiraishi, D. Tanabe, H. Morikawa, D. Barrett, S. albicans multidrug efflux pump MDR1 by its regulators Mrr1 and Cap1,
Matsumoto, K. Ohtomo, K. Maki, FR290581, a novel sordarin derivative: Antimicrob. Agents Chemother. 55 (2011) 2061–2066.
synthesis and antifungal activity, Bioorg. Med. Chem. Lett. 19 (2009) 1465– [108] D. Calabrese, J. Bille, D. Sanglard, A novel multidrug efflux transporter gene of
1468. the major facilitator superfamily from Candida albicans (FLU1) conferring
[81] D.W. Cleveland, K.F. Sullivan, Molecular biology and genetics of tubulin, resistance to fluconazole, Microbiology 146 (2000) 2743–2754.
Annu. Rev. Biochem. 54 (1985) 331–366. [109] N. Dunkel, T.T. Liu, K.S. Barker, R. Homayouni, J. Morschhäuser, P.D. Rogers, A
[82] A.E. Oxford, H. Raistrick, P. Simonart, Studies in the biochemistry of micro- gain-of-function mutation in the transcription factor Upc2p causes
organisms, Griseofulvin, C17H17O6Cl, a metabolic product of Penicillium upregulation of ergosterol biosynthesis genes and increased fluconazole
griseo-fulvum Dierckx, Biochem. J. 33 (1939) 240–248. resistance in a clinical Candida albicans isolate, Eukaryot. Cell 7 (2008) 1180–
[83] F.C. Odds, Antifungal agents: their diversity and increasing sophistication, 1190.
Mycologist 17 (2003) 51–55. [110] E. Snelders, W.J. Melchers, P.E. Verweij, Azole resistance in Aspergillus
[84] K. Gauwerky, C. Borelli, H.C. Korting, Targeting virulence: a new paradigm for fumigatus: a new challenge in the management of invasive aspergillosis?
antifungals, Drug Discov. Today 14 (2009) 214–222. Future Microbiol. 6 (2011) 335–347.
[85] J.W. Rippon, Medical Mycology: The Pathogenic Fungi and the Pathogenic [111] K.W. Henry et al., Upregulation of ERG genes in Candida species by azoles and
Actinomycetes, Saunders, Philadelphia, 1982. other sterol biosynthesis inhibitors, Antimicrob. Agents Chemother. 44
[86] Y. Miao, J.L. Tenor, D.L. Toffaletti, E.J. Washington, J. Liu, W.R. Shadrick, et al., (2000) 2693–2700.
Structures of trehalose-6-phosphate phosphatase from pathogenic fungi [112] T.C. White, Increased mRNA levels of ERG16, CDR, and MDR1 correlate with
reveal the mechanisms of substrate recognition and catalysis, Proc. Natl. increases in azole resistance in Candida albicans isolates from a patient
Acad. Sci. U.S.A. 113 (2016) 7148–7153, http://dx.doi.org/10.1073/ infected with human immunodeficiency virus, Antimicrob. Agents
pnas.1601774113. Chemother. 41 (1997) 1482–1487.
[87] M.F. Mabanglo, M.A. Hast, N.B. Lubock, H.W. Hellinga, L.S. Beese, Crystal [113] J.L. Lopez-Ribot, R.K. McAtee, L.N. Lee, W.R. Kirkpatrick, T.C. White, D.
structures of the fungal pathogen Aspergillus fumigatus protein Sanglard, T.F. Patterson, Distinct patterns of gene expression associated with
farnesyltransferase complexed with substrates and inhibitors reveal development of fluconazole resistance in serial Candida albicans isolates from
features for antifungal drug design, Protein Sci. 23 (2014) 289–301. human immunodeficiency virus-infected patients with oropharyngeal
[88] L.E. Cowen, S.D. Singh, J.R. Köhler, C. Collins, A.K. Zaas, W.A. Schell, et al., candidiasis, Antimicrob. Agents Chemother. 42 (1998) 2932–2937.
Harnessing Hsp90 function as a powerful, broadly effective therapeutic [114] J.C.O. Sardi, L. Scorzoni, T. Bernardi, A.M. Fusco-Almeida, M.J.S. Mendes
strategy for fungal infectious disease, Proc. Natl. Acad. Sci. U.S.A. 106 (2009) Giannini, Candida species: current epidemiology, pathogenicity, biofilm
2818–2823. formation, natural antifungal products and new therapeutic options, J. Med.
[89] P.A. Mann, C.A. McLellan, S. Koseoglu, Q. Si, E. Kuzmin, A. Flattery, et al., Microbiol. 62 (2013) 10–24.
Chemical genomics-based antifungal drug discovery: targeting [115] S. Fanning, A.P. Mitchell, Fungal biofilms, PLoS Pathol. 8 (2012) e1002585,
glycosylphosphatidylinositol (GPI) precursor biosynthesis, ACS Infect. Dis. 1 http://dx.doi.org/10.1371/journal.ppat.1002585.
(2015) 59–72. [116] J.V. Desai, A.P. Mitchell, D.R. Andes, Fungal biofilms, drug resistance, and
[90] M. Spitzer, E. Griffiths, K.M. Blakely, J. Wildenhain, L. Ejim, L. Rossi, et al., recurrent infection, Cold Spring Harb. Perspect. Med. 4 (2014) a019729.
Cross-species discovery of syncretic drug combinations that potentiate the [117] J. Nett, D. Andes, Candida albicans biofilm development, modeling a host–
antifungal fluconazole, Mol. Syst. Biol. 7 (2011) 499. pathogen interaction, Curr. Opin. Microbiol. 9 (2006) 340–345.

Please cite this article in press as: S. Campoy, J.L. Adrio, Antifungals, Biochem. Pharmacol. (2016), http://dx.doi.org/10.1016/j.bcp.2016.11.019
S. Campoy, J.L. Adrio / Biochemical Pharmacology xxx (2016) xxx–xxx 11

[118] J.E. Nett, H. Sanchez, M.T. Cain, K.M. Ross, D.R. Andes, Interface of Candida activity against cryptococcal meningitis with an improved formulation,
albicans biofilm matrix-associated drug resistance and cell wall integrity Mycoses 57 (2014) 42–43.
regulation, Eukaryot. Cell 10 (2011) 1660–1669. [133] F. Lamoth, B.D. Alexander, Antifungal activities of SCY-078 (MK-3118) and
[119] K.F. Mitchell, R. Zarnowski, H. Sanchez, J.A. Edward, E.L. Reinicke, J.E. Nett, standard antifungal agents against clinical non-Aspergillus mold isolates,
et al., Community participation in biofilm matrix assembly and function, Antimicrob. Agents Chemother. 59 (2015) 4308–4311.
Proc. Natl. Acad. Sci. U.S.A. 112 (2015) 4092–4097, http://dx.doi.org/10.1073/ [134] N.P. Wiederhold, L.K. Najvar, S. Matsumoto, R.A. Bocanegra, M.L. Herrera, B.L.
pnas.1421437112. Wickes, W.R. Kirkpatrick, T.F. Patterson, Efficacy of the investigational
[120] J. Bonhomme, C. d’Enfert, Candida albicans biofilms: building a echinocandin ASP9726 in a guinea pig model of invasive pulmonary
heterogeneous, drug-tolerant environment, Curr. Opin. Microbiol. 16 (2013) aspergillosis, Antimicrob. Agents Chemother. 59 (2015) 2875–2881.
398–403. [135] H. Morikawa, M. Tomishima, N. Kayakiri, T. Araki, D. Barrett, S. Akamatsu,
[121] D.M. Kuhn, T. George, J. Chandra, P.K. Mukherjee, M.A. Ghannoum, Antifungal et al., Synthesis and antifungal activity of ASP9726, a novel echinocandin
susceptibility of Candida biofilms: unique efficacy of amphotericin B lipid with potent Aspergillus hyphal growth inhibition, Bioorg. Med. Chem. Lett. 24
formulations and echinocandins, Antimicrob. Agents Chemother. 46 (2002) (2014) 1172–1175.
1773–1780. [136] T. Shibata, T. Takahashi, E. Yamada, A. Kimura, H. Nishikawa, H. Hayakawa,
[122] M. Montejo, Epidemiology of invasive fungal infection in solid organ et al., T-2307 causes collapse of mitochondrial membrane potential in yeast,
transplant, Rev. Iberoam. Micol. 28 (2011) 120–123 (in Spanish). Antimicrob. Agents Chemother. 56 (2012) 5892–5897.
[123] D. Sanglard, A.T. Coste, Activity of isavuconazole and other azoles against [137] J.D. Oliver, G.E.M. Sibley, N. Beckmann, K.S. Dobb, M.J. Slater, L. McEntee,
Candida clinical isolates and yeast model systems with known azole et al., F901318 represents a novel class of antifungal drug that inhibits
resistance mechanisms, Antimicrob. Agents Chemother. 60 (2015) 229–238. dihydroorotate dehydrogenase, Proc. Natl. Acad. Sci. U.S.A. 113 (2016)
[124] T.M. Diaz-Guerra, E. Mellado, M. Cuenca-Estrella, J.L. Rodriguez-Tudela, A 12809–12814, http://dx.doi.org/10.1073/pnas.1608304113.
point mutation in the 14a-sterol demethylase gene cyp51A contributes to [138] K. Koselny, J. Green, L. Favazzo, V.E. Glazier, L. DiDone, S. Ransford, D.J.
itraconazole resistance in Aspergillus fumigatus, Antimicrob. Agents Krysan, Antitumor/antifungal celecoxib derivative AR-12 is a non-nucleoside
Chemother. 47 (2003) 1120–1124. inhibitor of the ANL-family adenylating enzyme acetyl CoA synthetase, ACS
[125] D.S. Perlin, Resistance to echinocandin-class antifungal drugs, Drug Resist. Infect. Dis. 2 (2016) 268–280, http://dx.doi.org/10.1021/acsinfecdis.5b00134.
Update 10 (2007) 121–130. [139] M.C. Arendrup, R.H. Jensen, M. Cuenca-Estrella, In vitro activity of ASP2397
[126] G. Garcia-Effron, S. Park, D.S. Perlin, Correlating echinocandin MIC and kinetic against Aspergillus isolates with or without acquired azole resistance
inhibition of fks1 mutant glucan synthases for Candida albicans: implications mechanisms, Antimicrob. Agents Chemother. 60 (2016) 532–536.
for interpretive breakpoints, Antimicrob. Agents Chemother. 53 (2009) 112– [140] F. Peláez, A. Cabello, G. Platas, M.T. Díez, A. González del Val, A. Basilio, I.
122. Martán, et al., The discovery of enfumafungin, a novel antifungal compound
[127] P. Vandeputte, L. Pineau, G. Larcher, T. Noel, D. Brèthes, D. Chabasse, et al., produced by an endophytic Hormonema species biological activity and
Molecular mechanisms of resistance to 5-fluorocytosine in laboratory taxonomy of the producing organisms, Syst. Appl. Microbiol. 23 (2000) 333–
mutants of Candida glabrata, Mycopathologia 171 (2011) 11–21. 343.
[128] B.M. Vincent, A.K. Lancaster, R. Scherz-Shouval, L. Whitesell, S. Lindquist, [141] C. Jiménez-Ortigosa, P. Paderu, M.R. Motyl, D.S. Perlin, Enfumafungin
Fitness trade-offs restrict the evolution of resistance to amphotericin B, PLoS derivative MK-3118 shows increased in vitro potency against clinical
Biol. 11 (2013) e1001692. echinocandin-resistant Candida species and Aspergillus species isolates,
[129] J.A. Maertens, I.I. Raad, K.A. Marr, T.F. Patterson, D.P. Kontoyiannis, O.A. Antimicrob. Agents Chemother. 58 (2014) 1248–1251.
Cornely, Isavuconazole versus voriconazole for primary treatment of invasive [142] J.M. Apgar, R.R. Wilkening, M.L. Greenlee, J.M. Balkovec, A.M. Flattery, G.K.
mould disease caused by Aspergillus and other filamentous fungi (SECURE): a Abruzzo, et al., Novel orally active inhibitors of b-1,3-glucan synthesis
phase 3, randomised-controlled, non-inferiority trial, Lancet 387 (2016) 760– derived from enfumafungin, Bioorg. Med. Chem. Lett. 25 (2015) 5813–5818.
769. [143] N.P. Wiederhold, L.K. Najvar, A.W. Fothergill, D.I. McCarthy, R. Bocanegra, M.
[130] A.G.S. Warrilow, C.M. Hull, J.E. Parker, E.P. Garvey, W.J. Hoekstra, W.R. Moore, Olivo, et al., The investigational agent E1210 is effective in treatment of
et al., The clinical candidate VT-1161 is a highly potent inhibitor of Candida experimental invasive candidiasis caused by resistant Candida albicans,
albicans CYP51 but fails to bind the human enzyme, Antimicrob. Agents Antimicrob. Agents Chemother. 59 (2015) 690–692.
Chemother. 58 (2014) 7121–7127. [144] R.H. Jensen, K.M.T. Astvad, L.V. Silva, D. Sanglard, R. Jørgensen, K.F. Nielsen,
[131] E.P. Garvey, W.J. Hoekstra, R.J. Schotzinger, J.D. Sobel, E.A. Lilly, P.L. Fidel, et al., Stepwise emergence of azole, echinocandin and amphotericin B
Efficacy of the clinical agent VT-1161 against fluconazole-sensitive and - multidrug resistance in vivo in Candida albicans orchestrated by multiple
resistant Candida albicans in a murine model of vaginal Candidiasis, genetic alterations, J. Antimicrob. Chemother. 70 (9) (2015) 2551–2555,
Antimicrob. Agents Chemother. 59 (2015) 5567–5573. http://dx.doi.org/10.1093/jac/dkv140.
[132] L.K. Najvar, N.P. Wiederhold, A. Alimardanov, J. Cradock, X. Xu, M. Behnke,
et al., The novel fungal Cyp51 inhibitor VT-1129 demonstrates potent in vivo

Please cite this article in press as: S. Campoy, J.L. Adrio, Antifungals, Biochem. Pharmacol. (2016), http://dx.doi.org/10.1016/j.bcp.2016.11.019

You might also like