SlideShare a Scribd company logo
1 of 21
Download to read offline
Contents lists available at ScienceDirect
Bioorganic Chemistry
journal homepage: www.elsevier.com/locate/bioorg
History of the development of antifungal azoles: A review on structures,
SAR, and mechanism of action
Mohammad Shafieia
, Lee Peytonb
, Mehrnoosh Hashemzadehc,⁎
, Alireza Foroumadia,⁎
a
Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
b
Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
c
University of Arizona college of medicine Phoenix and Pima college, Tucson, AZ, USA
A R T I C L E I N F O
Keywords:
Invasive fungal infection
Lanosterol 14α-demethylase
Azole
Structure activity relationship
Drug development
Molecular hybrids
A B S T R A C T
With the increasing risk of invasive and life threating fungal infections, there is now a great concern regarding
the lower discovery rate of antifungal drugs in comparison to antimicrobial agents. Drugs conventionally used in
clinics are not adequate enough to combat the increasing fungal infections, especially fungal forms resistant to
fluconazole. Among the limited antifungal agents in clinics, azoles have the largest number of drug candidates in
clinical trials and are partly marketed due to the particular focus of pharmaceutical companies and medicinal
scientific centers. With the rise in the number of papers on azole antifungal design and discovery, a more in-
depth understanding the most recent and authentic information about this class of drugs might be beneficial. To
this end, we for the first time summarized the state-of-the-art information about azole drugs, with a specific focus
on those in the pipelines of pharmaceutical companies, into four generations with regard to their structural
similarity. More importantly, this review highlights information on the structure activity relationship (SAR),
target description, hybrid antifungal agents as possible future generation, and other useful issues to streamline
research towards designing new efficient azole antifungal structures in future.
1. Introduction
1.1. Fungal infection
The Fungi kingdom is one of the most diverse organismal kingdoms
on the planet. Unlike the bacteria, fungi have eukaryotic cellular
functions, making them more closely related to human than bacteria
[1]. Out of more than two million fungi species, about 600 types are
known as human fungal pathogens, and only 3–4% of these species
account for > 99% of invasive fungal infection (IFI) [2]. These types of
fungal infections are often life-threatening and have a higher mortality
rate in comparison to superficial fungal infections, which are common
in human beings [3–5].
A particularly susceptible population for the development of IFIs are
individuals with compromised immune function such as those with
organ transplants and AIDS (acquired immune deficiency syndrome) as
well as those who are on prolonged immunosuppressive medications
such as corticosteroids. The patients with antibiotics, cancer che­
motherapies, and the ones in intensive care units (ICU) are also at
greater risk to develop IFI [4]. This sheds some light on the opportu­
nistic nature of fungal infection [6]. Over the last 30 years, IFIs in­
creased with the rising number of the vulnerable population and an­
nually caused 1–2 million deaths. The mortality rate was much greater
than that of malaria or tuberculosis [7–9]. > 90% of the deaths are
attributed to Candida and aspergillus species [10]. Candida species, in­
cluding Candida albicans, Candida parapsilosis, Candida krusei, Candida
glabrata, and Candida tropicalis have emerged as invasive candidiasis,
whose mortality rate is increased up to 75% as they usually affect re­
spiratory, gastrointestinal, skin, and urogenital tracts [11–13]. More­
over, aspergillosis along with aspergillus spp. such as fumigatus, flavus,
niger, terreus, and parasiticus mainly affect external ear, lungs, eye, and
brain, with a mortality rate of 50–90% [14].
Although Fungi infection is the first scientifically reported or­
ganism-derived disease, prior to bacterial infections [15], antifungal
drug discovery has mightily lagged behind antibacterial chemotherapy,
resulting in one of the main problems with targeting fungal species, i.e.
the eukaryotic nature of fungi makes them intrinsically difficult to be
targeted without the probable toxic effects of antifungal drugs in
https://doi.org/10.1016/j.bioorg.2020.104240
Received 9 June 2020; Received in revised form 17 July 2020; Accepted 11 August 2020
⁎
Corresponding authors at: University of Arizona college of medicine Phoenix and Pima college, Tucson, Arizona, USA. (Mehrnoosh Hashemzadeh). Department of
Medicinal Chemistry, Faculty of Pharmacy & The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran (Alireza
Foroumadi).
E-mail addresses: mhashemz1@yahoo.com (M. Hashemzadeh), aforoumadi@yahoo.com (A. Foroumadi).
Bioorganic Chemistry 104 (2020) 104240
Available online 28 August 2020
0045-2068/ © 2020 Elsevier Inc. All rights reserved.
T
human cells [15]. Accordingly, the development of selective antifungal
drugs with lower side-effect is a hotbed of research among medicinal
chemists [16].
1.2. Current medications
Currently, there are several well-known mechanisms to treat sys­
temic and superficial fungal infections using roughly 40 branded anti­
fungal drugs in the market and the other drug candidates in clinical
trials, which adopt new or convenient mechanisms to target fungi
(Fig. 1) [2,17–20].
Interestingly, the previous studies have introduced some new tar­
gets to combat fungal infections, hoping that the emerging strategies
may yield the treatment for resistant fungal infections (Fig. 2) [21–24].
Despite new research and development in this field, there is still no
new approved drug for the more recent strategies. As shown in Figs. 1-
14-α demethylase inhibitors (azoles) are the most important and the
most approved clinical candidate drugs.
According to previous studies, classified azoles cover several char­
acteristics of the most potent antifungals. Over the past half-century,
this family has had a significant effect on controlling invasive and su­
perficial fungal infections. Their newer generation further increased
highly desired properties such as safety, lower risk of drug-drug inter­
actions, proper pharmacokinetics, and improved activity spectrum
[25–27].
With the large number of azole antifungals in the clinics and in
experimental medicinal chemistry institutions, keeping track of pro­
mising molecules that are under development is challenging for the
researchers in the field. In this paper, we provide a comprehensive
review and an update of all approved and clinical candidates of azole
antifungal drugs, as azole pedigree in regard to their structural simi­
larities. Herein, the description of the target, salient medicinal chem­
istry points regarding azole structures (SAR), and the selectivity of these
structures for the concerned target are discussed. Furthermore, hybrid
antifungal agents as a prospective future generation are introduced to
pave the way for developing new efficient azole antifungal structures in
the future.
2. Azole family
2.1. History and pedigree
The first report on the antifungal activity of an azole compound was
presented by Woolley (1944), a serendipitous discovery that declared
the antimycotic effect of benzimidazole moiety for the first time [28].
Afterwards, several other researchers indicated other azole antifungal
structures such phenethylimidazole [29] and substituted benzimida­
zoles [30]; however, none of these reports led to the development a
drug until 1958, when chlormidazole (1, Fig. 3) was introduced, the
first azole drug marketed by Chemie Grünenthal. Accordingly, chlor­
midazole was the pioneer of enormous research on azole antifungal
activities. Currently, there are about 40 azole-containing drugs and
drug candidates, which could be classified into more than three gen­
erations [15,31,32]. This field of study is still of great interest.
2.1.1. First generation
The first generation of azole family emerged with the introduction
of three compounds: clotrimazole (13) from Bayer AG in 1969,
Janssen’s miconazole (2) at the same time, and later econazole (3) in
1974. These three drugs were launched as topical antifungal agents
making azoles an indispensable asset in treating fungal infections[15].
Following the discovery of these early structures, several analog com­
pounds are marketed, which can be categorized into the first-generation
group due to their similar physiochemical properties and indications in
treating superficial fungal infections such as dermatomycoses, tinea
versicolor, cutaneous, and vaginal candidiasis as well as topical for­
mulations. Considering their structures, they have similar imidazole-
contained backbone and could be further sub-divided into three cate­
gories: miconazole-based structures, clotrimazole- based structures, and
vinyl-imidazole derivations, as shown in Fig. 4.
Fig. 1. Antifungal drugs. the number of approved and clinical trial agents are summarized.
M. Shafiei, et al. Bioorganic Chemistry 104 (2020) 104240
2
Miconazole (2) has well-documented activity in the treatment of
oropharyngeal candidiasis as well as vulvovaginal candidiasis, tinea
corporis, tinea pedis, and tinea cruris [33]. It also has a fungicidal ac­
tivity via direct membrane damage. Clinically, miconazole adminis­
tration routes are topically or intravenously; however, miconazole in­
fusion is also associated with adverse reactions such as nausea, fever,
and more importantly cardiotoxicity when rapidly infusing that was
due to a formulation containing Cremophor® EL because of its low so­
lubility [34]. All the miconazole analogs are derived from a key inter­
mediate, 2-(imidazolyl)-acetophenone. The only exception is butoco­
nazole (11), which is derived from 4-(4-chlorophenyl)-1-(1H-imidazole-
1-yl) butan-2-ol structure.
According to FDA, clotrimazole (13) is safe and effective for the
prevention and treatment of oropharyngeal, vulvovaginal, and
cutaneous candidiasis and the other superficial dermatologic infections
[35].
Clotrimazole (13) synthesis was reported three years after its de­
velopment. Benzophenone derivations could be used for clotrimazole
(13) and bifonazole (14), and dibenzosuberone for eberconazole (15)
synthesis. Becliconazole (16) is a new clotrimazole-like compound,
which is recently entered into the clinical trials from Menarini
Company.
Luliconazole (21), the newest first-generation azole approved by
FDA, contains a unique vinyl-imidazole structure. It has a broad-spec­
trum topical effect against dermatophytes such as Trichophyton rubrum,
Microsporum gypseum and Epidermophyt on floccosum [36]. In this group,
TS-80 (22) entered into phase-I clinical trial from Shikoku company and
revealed an activity against cutaneous fungal infections; however, this
trial has not yet been completed.
2.1.2. Second generation
For second generation azole antifungals, there are three main
changes in the structure of the compounds which resulted in many
improvements in terms of the safety, spectrum of action, and pharma­
cokinetics of azole drugs. It should be noted that following the second
generation of azoles, subsequent generations are just modifications and
analogs of these structures.
In this regard, the main changes are evident in Janssen's (1981)
antifungal ketoconazole (23), the first drug containing a heterocyclic
dioxolane functional group, replacing the ketone with a new long-tail
Fig. 2. New fungal cell targets.
(1)
Fig. 3. Structure of chlormidazole (1).
M. Shafiei, et al. Bioorganic Chemistry 104 (2020) 104240
3
structure; however, the imidazole moiety remained similar to its pre­
decessor. This was the first orally active, broad-spectrum azole to treat
systemic mycoses [37].
Although ketoconazole (23) has several drawbacks resulting in a
black box warning from the US FDA, it was preferred to amphotericin B
(AmB) for non-life threatening systemic fungal infections until 1990
[38]. One of the major problems with ketoconazole (23) is hepato­
toxicity caused by CYP450 inhibitions, indicating the imidazole-con­
tained lower selectivity for fungal compared to mammalian P450 en­
zymes. The other prevailing problems are bioavailability and
pharmacokinetic interactions, which finally led to removal of this drug
from the FDA list of approved medication to treat systemic mycosis in
2013 [39].
The third and the most important breakthrough in the development
of azole structure was the introduction of a triazole ring replacing the
bygone imidazole moiety in the azole structures. Triazole was first used
along with fluconazole (24) developed by Pfizer (1989) [40]. The su­
perior advantages of fluconazole (24) were selectivity for fungal CYP
enzyme over mammalian, a larger spectrum of action, increased
metabolic stability, lower protein binding, enhanced bioavailability,
greater water solubility to be used as intravenous indications, high CNS
penetration, and lower side-effects [41–43]. Interestingly to achieve
higher IV bolus administration of fluconazole (24) in clinics, even
greater solubility was necessary. This problem was solved using a
phosphate ester of fluconazole (24) which was approved in 2003 [44].
Fosfluconazole (28) could be active with the action of alkaline phos­
phatase enzymes [45].
The discovery of fluconazole (24) is one of the most interesting
medicinal chemistry studies on the modification of a structure to
achieve desirable properties (Fig. 5) [46]. In its synthesis, an epoxide
ring (Fig. 6A) is the key intermediate achieved by well-known Corey-
chaykovsky epoxidation with trimethyl sulfoxonium iodide (TMSI) re­
agent employed as the ring closer [47–50].
At the same period, another important triazole-based structures
improved by Janssen pharmaceuticals were terconazole (25) and itra­
conazole (26) [15], both of which had a long-tail structure to similar
that of ketoconazole (23) as well as similar low bioavailability. They
have several side-effects, including cardiotoxicity. Although they have a
Fig. 4. First generation azole structures: A: miconazole (2) analogs, B: clotrimazole (13) analogs, C: Vinyl-imidazole structures.
M. Shafiei, et al. Bioorganic Chemistry 104 (2020) 104240
4
broader spectrum of antifungal activity in comparison to ketoconazole
(23) and fluconazole (24), they couldn’t be an alternative to fluconazole
(24) in many cases [51–53]. Itraconazole (26), however, was preferred
to ketoconazole (23) in the treatment of mycoses, Blastomycosis and
invasive aspergillosis [54,55]. In terms of synthesis, itraconazole (26)
and similar structures utilize a common route through dioxolane in­
termediate formation at four major steps (Fig. 6B) to form the targeted
compound [37].(See Table 1)
When the first-generation structures were compared to the second-
generation (Fig. 7), the latter generation improved upon the spectrum
of activity and added routes of administration; however, there were still
challenges regarding pharmacokinetic and safety profiles. Conse­
quently, the third generation of azole drugs were the improved struc­
tures of the second generation and could be sub-divided into two groups
of fluconazole (24) and itraconazole (26)-like structures. Table 2 shows
the major physiochemical and pharmacokinetic properties of the main
second-generation azole drugs [45,56,57].
2.1.3. Third generation
With the increasing concerns about the high incidence of fungal
infections, new structures emerged as the third-generation azoles or
triazole generation. These drugs are improved structures of the second-
generation antifungals divided into two groups of fluconazole (24) and
itraconazole (26) like structures (Fig. 8). In both of these groups of the
third generation, major medicinal chemical efforts were made to im­
prove pharmacokinetic properties and safety profile, extend the spec­
trum of activity, and, more importantly, design a structure to combat
resistant types of fungal infections. In Table 3, major physiochemical
Fig. 4. (continued)
M. Shafiei, et al. Bioorganic Chemistry 104 (2020) 104240
5
and pharmacokinetic properties of several third generation drugs are
listed [58–67].
Since fluconazole (24) is inactive against molds such as Aspergillus
spp and increases the resistance of Candida spp, in 2002, the FDA ap­
proved voriconazole (30) produced by Pfizer with a fluoropyrimidine
ring instead of the second triazole of fluconazole (24) [68]. This
structural difference has made the compound > 30 fold more potent for
C. albicans, 20 fold more potent for C. glabrata, and completely active
against all Aspergillus spp [69].
In 2004, the Schering Plough Research Institute working on a new
lead, SCH51048, observed that antifungal activity was higher than the
hypothesized concentration, suggesting the presence of an active
metabolite. Following LC-MS/MS analysis, a hydroxylated metabolite
was discovered. By using this as a lead and after synthesis of all side-
chain monohydroxylated diastereomers, posaconazole (31, SCH56592)
was discovered as a new triazole antifungal agent, providing a prime
example of active metabolite-based drug design (Fig. 9) [70,71].
Posaconazole (31) showed a broader spectrum of activity against all
fungus organisms covered by other new broad-spectrum antifungal
drugs. Moreover, it had an additional activity against Zygomycetes,
Scedosporium spp, and also was effective in the treatment of CNS fungal
infections and cryptococcal meningitis [72].
The FDA approved posaconazole (31) for prophylaxis of invasive
Aspergillus or Candida infections in hematopoietic stem cell
Fig. 4. (continued)
Fig. 5. Fluconazole (24) structure optimizations.
M. Shafiei, et al. Bioorganic Chemistry 104 (2020) 104240
6
transplantation (HSCT) recipients with graft versus host disease
(GVHD), as well as in patients with hematologic malignancy with
prolonged neutropenia and oropharyngeal candidiasis [73–75].
Efinaconazole (32), as a new topical triazole antifungal for the
treatment of mild to moderate onychomycosis, received its first global
approval on October 3, 2013 in Canada by Valeant Pharmaceuticals
International. This compound has a better safety profile compared with
the oral treatment of onychomycosis [76].
In cases of intravenous formulations of posaconazole (31), vor­
iconazole (30), and itraconazole (26), cyclodextrin is added to increase
solubility, which is of concern due to potential nephrotoxicity [77].
This concern was remedied by the medicinal chemistry work of Basilea
Pharmaceuticals and Astellas Pharma. Isavuconazonuim sulfate, a
water-soluble prodrug of isavuconazole (33) containing a 1-[N-methyl-
N-[3-[2-(methylamino)acetoxymethyl]pyridyn-2-yl]amino carbony­
loxy] ethyl substituent, easily converts to its active form by the action of
plasma esterase (Fig. 10). Therefore, the oral and intravenous for­
mulation no longer needs cyclodextrin for enhanced solubility [78,79].
(See Fig. 11)
As a broad-spectrum azole drug, isavuconazole (33) has an anti­
fungal activity profile similar to that of voriconazole (30). In 2014, the
FDA approved it for treatment of invasive aspergillosis [80].
Another example of triazole antifungal in clinical trials is ravuco­
nazole (34), with a slight difference in structure compared to
isavuconazole (33), but with a similar antifungal profile to that of
voriconazole (30). Ravuconazole (34) is active against fluconazole (24)
and itraconazole (26) resistance infections. It was discontinued from
phase II for aspergilusis and candidiasis in August 2007, and was re­
placed with fosravuconazole bis(L-lysine) created by Bristol-Myers
Squibb. The first global approval of this prodrug of ravuconazole (34)
with the brand name of NAILIN® as 100 mg capsules was for onycho­
mycosis in Japan on July 27, 2018 [81].
Albaconazole (35) with a 7-chloro-quinazoline-4-one structure
bears more potent antifungal in vitro activity than Amphotericin B.
Furthermore, a wide spectrum of action and favorable pharmacokinetic
with safety profile [82] was originated by Grupo Uriach. In 2013, Al­
lergan (Actavis Inc) picked up the worldwide rights for albaconazole
(35) from Palau Pharma S.A.
Other new compounds of this generation are also currently in phase-
III, such as genaconazole (36) and iodiconazole (37), phase II such as
saperconazole (39), ICI-D0870 (42) and UR-9751 (41), and phase I such
as embeconazole (38), DUP-860 (40), SS-750 (44), SSY-726 (46) and
PC-945 (47). Moreover, some new structures like UR-9660 (43) and
SDZ-89–485 (45) are in preclinical stages [18–20,83].
Many triazole alcohol derivations have been investigated and sev­
eral new and known scaffolds incorporated into their structures are
assessed in terms of their antifungal activity [84–86]. This new tech­
nique for antifungal agent discovery is highly growing in medicinal
Fig. 6. Second generation azole drugs intermediates synthesis, A: The intermediate of Iitraconazole (dioxalene) synthesis roots. B: Fluconazole’s intermediate
(epoxide) synthesis root.
M. Shafiei, et al. Bioorganic Chemistry 104 (2020) 104240
7
chemistry research [87–95].
2.1.4. Fourth generation
In the previous generations, new structures successfully emerged
and solved the resistance, potency, the spectrum of action, and phar­
macokinetics problems of azole family. One of the main problems,
known as the ‘Achilles heel’ for azoles, is the drug-drug interactions
caused by probable human CYP3A4 enzyme inhibition [33]. This pro­
blem is a concern since many patients suffer from complicated
conditions such as AIDS, cancer, and impaired immune function and
need polypharmacy. The metal binding region in azole structures is the
main part of the inhibitory effect as for human CYP3A4 inhibition.
Following the replacement of imidazole with triazole ring, the se­
lectivity increases, even though, the problem still exists. Successful ef­
forts have been made in Viamet Pharmaceuticals, Inc. (Durham, NC,
USA). Using an appropriate medicinal chemistry strategy, they in­
troduced three novel azole structures (Fig. 10) with high selectivity for
fungal CYP51 and increased potency and desirable pharmacokinetics
and physiochemical properties (Table. 4), in comparison to the other
structures [96–98]. Replacement of high-avid metal binding and more
potent triazole ring with less-avid metal binding and less potent tetra­
zole ring and the compensation of low potency with modification of
side-chain were performed to have the more selective and potent
fourth-generation of azole structures [99,100].
They used voriconazole (30) for its lower anti-target (CYP3A4) af­
finity compare to the other azoles, as a leading compound to design new
compounds. Quilseconazole (48, VT-1129) is now in phase I clinical
trial in cryptococcal meningitis. It has had a promising efficacy in the
murine model and increased one-month survival, compared to fluco­
nazole (24). In Jun 2016, VT-1129 (48) received Fast Track orphan
drug designation for the treatment of cryptococcal meningitis [PO] (in
volunteers) in the USA [101,102].
Table 1
First generation azole information’s about important physiochemical proper­
ties.
Generic name Physiochemical properties
CLogP1
tPSA2
LogS3
Becliconazole 4.83 24.83 −5.94
Bifonazole 4.74 15.6 −6.28
Butoconazole 6.86 15.6 −7.56
Clotrimazole 5 15.6 −6.97
Croconazole 4.5 24.83 −5.68
Dapaconazole 5.26 24.83 −6.64
Eberconazole 4.97 15.6 −6.1
Econazole 5.1 24.83 −6.39
Fenticonazole 6.72 24.83 −8.23
Flutrimazole 4.57 15.6 −6.75
Isoconazole 5.8 24.83 −7.08
Luliconazole 3.49 39.39 −5.54
Miconazole 5.8 24.83 −7.09
Neticonazole 4.24 24.83 −5.48
Omoconazole 6.06 34.06 −7.44
Oxiconazole 6.46 37.19 −7.69
Sertaconazole 6.16 24.83 −7.45
Sulconazole 6.27 15.6 −7.02
Tioconazole 4.78 24.83 −5.76
Ts-80 5.52 32.6 −6.98
Zinoconazole 5.08 39.9 −6.198
1. Calculated octanol–water partition coefficient, 2. Topological polar surface
area, 3. Aqueous solubility
Fig. 7. Second generation azole drugs structures.
Table 2
Major physiochemical and pharmacokinetic properties of important second
generation azole drugs.
Generic name Physiochemical properties Pharmacokinetic properties
CLogP1
tPSA2
LogS3
PPB (%)a
Half-life
(h)
Vd (L/
Kg)b
Fluconazole −0.44 76.15 −2.127 12 31 0.7
Itraconazole 5.98 98.04 −9.752 > 95 64 11
Ketoconazole 3.63 66.84 −6.421 > 95 8 No data
1. Calculated octanol–water partition coefficient, 2. Topological polar surface
area, 3. Aqueous solubility a: plasma protein binding, b: volume of distribution
M. Shafiei, et al. Bioorganic Chemistry 104 (2020) 104240
8
Oteseconazole (49, VT-1161) another tetrazole compound after
successful phase III trial for vulvovaginal candidiasis in March 2019
(PO, Capsule) (NCT03840616), the Mycovia Pharmaceuticals and
Jiangsu Hengrui Medicine have teamed up to develop and market the
first FDA approved drug for vulvovaginal candidiasis [103,104]. VT-
1598 (50), with the broadest spectrum of antifungal activity among
tetrazoles, is the most recent compound from the tetrazole group [105],
whose important anti-coccidioidomycosis effect (in vitro MIC:
0.06–0.5 mg/L) import this compound to preclinical development in
USA (PO) [106]. Another multidrug resistance fungal spices, Candida
auris, is also affected by VT-1598 (50) with MIC of 0.03–8 mg/L in vitro
[107].
Following the efforts of Viamet pharmaceuticals, the significance of
side-chain optimization in increasing the potency and metal-binding
Fig. 8. Third generation azole drug structures (blue structures derived from fluconazole and red structures derive from itraconazole). (For interpretation of the
references to colour in this figure legend, the reader is referred to the web version of this article.)
M. Shafiei, et al. Bioorganic Chemistry 104 (2020) 104240
9
group tetrazole in having better selectivity became obvious. This new
family and all other new structures in clinical trials showed that an old
mechanism could be a new hope.
2.1.5. Hybrid structures as future azole-generation
The hit to lead process and further development to achieve a drug
candidate is time and cost consuming [108]. Classical combinatorial
chemistry or modern virtual screening approaches have not yet brought
a breakthrough in the discovery of novel drug candidates [109]. In
order to obtain new antifungal drugs that are affordable and able to
avoid the emergence of resistant strains, hybrid drug discovery comes
to mind. Molecular hybrids of biologically active structures are now
gaining momentum worldwide and may be a key tool in the arsenal of
medicinal chemistry efforts for drug discovery [110]. Hybrid or dual-
acting molecules are defined as combination of two or more chemical
scaffolds that act at different targets to create more specific and
Fig. 8. (continued)
M. Shafiei, et al. Bioorganic Chemistry 104 (2020) 104240
10
powerful drugs [111].
Fluconazole (24) as a favorable antifungal drug due to its worthy in
vivo efficacy and pharmacokinetic property, has been attracted for
hybrid drug discovery. Fluconazole analogues obtained by facile
replacing one triazole ring by other potential antifungal scaffolds di­
rectly or by using suitable linkers [112]. This new way to design
structures makes fluconazole hybrids potential future generation of
azole antifungals.
The triazole nucleus especially 1,2,3-triazole ring has been utilized
as a linker in various antifungal drug hybridizations by means of click
reaction [113]. Several research groups introduced 1,2,3-triazole ring
in the side chain of azole structure to design hybrid structures and
survey antifungal effects [90,114–118](Fig. 15). This motif act not only
as a linker, but also interacts efficiently with active site of CYP51
providing in many cases improved physiochemical properties of struc­
tures [119].
Recently medicinal chemists have made considerable efforts to de­
sign potent hybrid antifungal structures of fluconazole, some of which
are summarized (Fig. 12).
Natural products are a dominant source of biologically active
structures and hybridization of these structures with fluconazole can
lead to potent drugs [108]. Zhang et al, [89] report design and synthesis
of novel series of carbazole-triazole conjugates. Carbazole derivations
as naturally occurring phytochemicals are known for pi-conjugated
Table 3
Major physiochemical and pharmacokinetic properties of several 3rd genera­
tion azole drugs.
Generic name Physiochemical properties Pharmacokinetic properties
CLogP1
tPSA2
LogS3
PPB (%)a
Half-life (h) Vd
(L/
Kg)b
Voriconazole 0.52 72.91 −2.72 58 6 4.6
Posaconazole 4.1 109.04 −8.82 > 95 25–31 428
Efinaconazole 2.14 51.43 −2.97 – 29.9 –
Isavuconazole 2.68 84.34 −5.19 98 56–77 6.5
Ravuconazole 2.68 84.34 −5.2 98 76–202 10.8
Albaconazole 2.14 80.86 −4.72 98 30–56 Very
large
1. Calculated octanol–water partition coefficient, 2. Topological polar surface
area, 3. Aqueous solubility a: plasma protein binding, b: volume of distribution
Fig. 9. Active metabolite of early lead SCH51048, posaconazole (31, SCH 56592).
M. Shafiei, et al. Bioorganic Chemistry 104 (2020) 104240
11
motifs that could inhibit topoisomerase, intercalate with DNA, and also
disrupt the integrity of the microorganism membrane [120]. Their ef­
forts led to compound A (Fig. 12) with MIC = 2–32 µg/mL against most
of the tested fungal strains. Compound A also, intercalates into DNA to
possibly block DNA reproduction. Elias et al [121] in an effort for
synthesis new azole hybrids with coumarin and quinolone scaffold
showed that their coumarin hybrids antifungals (compound B, Fig. 12)
could penetrate into endoplasmic reticulum of fungal cells where, true
target CYP51 generally exist in. This potentially can enhance the effi­
cacy of azole antifungal drugs.
Combination of known antifungal, antimicrobial, and anticancer
drugs to azole structures are also among high interest. This hybridiza­
tion will help to combat resistance strains due to the possibly different
mechanism of actions. Experiments conducted by Sheng et al, [122]
elucidate design and synthesis of the first CYP51/HDAC dual inhibitors.
They successfully developed potent antifungals agents with activity
against azole-resistant clinical isolates. The HDAC family of proteins are
key enzymes involved in gene regulation, cell proliferation, and are
known anticancer targets. Vorinostat (SAHA) (Fig. 12) an HDAC in­
hibitor, was previously confirmed to possess synergistic effects with
fluconazole (24) against azole-resistant strains. Inspired by these find­
ings, new anti-fungal dual inhibitor hybrids emerged. Compound C,
with MIC = 0.25–0.5 µg/mL and potent in vivo efficacy against azole
resistance candidiasis was introduced as the best CYP51/HDAC hybrid.
Before this, they also designed a hybrid of rosiglitazone and other de­
rivations of thiazolidinedione with fluconazole (Fig. 12) and
investigated anti-fungal activities. Their target compound D, with MIC
values against C. albicans in the range of 0.03–0.15 µM, proved the
efficiency of dual inhibitors in antifungal drug discovery [111].
Another team, studied the antifungal activity of 5-flucytosine
combination with fluconazole (123, Fig. 12). Two potent antifungals
combined in one molecule. Their efforts led to compound E with
MIC = 0.008 and 0.02 mM against C. albicans ATCC 90,023 and clinical
resistant strain C. albicans respectively. They also showed that new
hybrids could permeate membrane of C. albicans and also could inter­
calate into calf thymus DNA helix and block DNA replication by making
a steady supramolecular complex [123].
2.2. Mechanism of inhibition
The recognition of a bottleneck for efficient antifungal drug dis­
coveries based on the eukaryotic nature of fungi and its similarities with
the human is necessitous. Similar to most of the other drugs, the be­
ginning of azole class was by chance, and none of the antifungal drugs
were discovered using rational target-based drug design strategy.
One of the main targets of small molecules effective in combating
fungal infection is cell membrane and the biosynthesis of ergosterol, a
derivative of mammalian cholesterol. Ergosterol has several functions
in fungi, the important of which is its role in cellular proliferation. To
this end, a planar a-face (no methyl group at C14) structure and a
double bond in C5-6 are required to have these structural character­
istics. There are several important enzymes in the biosynthesis pathway
Fig. 10. Activation of Isavuconazonium sulfate prodrug to its active drug.
M. Shafiei, et al. Bioorganic Chemistry 104 (2020) 104240
12
[124], one of the predominant enzyme in required is CYP51 (Lanestrol-
14α-demethylase) (ERG-11) from cytochrome P450 family, which cat­
alyzes an oxidative removal of lanosterol, 14-methyl group and leads to
a 14–15 double bond (Fig. 13) [125]. In the active site of CYP51, an
iron-containing porphyrin provides oxygen for this first oxygen-de­
pendent reaction in the ergosterol biosynthesis. Recently, several pa­
pers fully-described the mechanistic details of CYP51 action [126–129].
Furthermore, a recent study showed that CYP51 might be involved
in distinct process of eluding human immune system, a finding that will
likely grow with the increase with the growth of research. [130]. Thus,
accumulation of methylated sterols would cause a fungistatic effect in
yeast and fungicidal effect in molds in most of the azoles, except in high
doses, and voriconazole (30) and itraconazole (26) (they are considered
to have fungicidal inhibition). Pertaining to immunocompromised pa­
tients, the eradication of the fungal infection is of essence and fungi­
cidal effects in therapeutic doses are highly beneficial [53].
Thus, knowing the enzyme structure is crucial for efficient
inhibition of CYP51 and designing selective and potent inhibitors.
Elucidation of the CYP51 structural information was challenging before
obtaining a complex of CYP51 orthologue from prokaryote myco­
bacterium tuberculosis (carrying about a 33–35% sequence homology to
that fungal) with fluconazole (24). The reason is that purification and
crystallization of a highly hydrophobic membrane-bound protein is
arduous.
Information from other CYP51 orthologues is not useful. For ex­
ample, VFV, as a potent anti T. cruzei drug candidate, today, has a weak
inhibitory effect on fungal CYP51. Moreover, VNI, as another anti-
Chagas disease drug candidate with an important H-bond interaction of
its carboxamide group with the T. cruzei active site, does not form any
H-bond in fungal active site protein [131–133]. However, biochemistry
works and X-ray crystallographic studies have recently developed a
protein expressed in some organisms like Escherichia coli, S. cerevisiae,
C. albicans, and C. glabrata and also in other important species
[129,134].
Fig. 11. Fourth generation azole drugs structures derive from voriconazole.
Table 4
Major physiochemical and pharmacokinetic properties of 4th generation azole drugs.
Generic name Physiochemical properties Pharmacokinetic properties
CLogP1
tPSA2
LogS3
PPB (%)a
Half-life (h) Vd (L/Kg)b
Quilseconazole 4.99 82.14 −6.59 No data > 6 days No data
Oteseconazole 4.67 82.14 −6.64 No data > 48 1.4
VT-1598 5.73 105.93 −7.91 > 95 22 No data
1. Calculated octanol–water partition coefficient, 2. Topological polar surface area, 3. Aqueous solubility a: plasma protein binding, b: volume of distribution
M. Shafiei, et al. Bioorganic Chemistry 104 (2020) 104240
13
From many studies of azole binding site interactions in the ligand-
binding pocket of CYP51 in different species reported by diverse
groups, several common interactions have emerged with conserved
amino acids in the active site. The most conserved domain is heme-
binding pocket, and also motifs that donate to set the heme into the
core structure and some other residues on the surface of the active site
[135]. Recent biochemistry studies declare that the ligand-binding
pocket (LBP) in different species such as S. cerevisiae, C. glabrata, and C.
albicans has similarities with the structural and chemical characteristics
of lanosterol demethylase protein [135]. Azole drugs inhibit the en­
zyme reaction by non-competitive reversible interaction with the heme
and prevent accessing of protons to the active site. There are several
similar interactions with other residues. For example, structures have
shown that C. albicans CYP51 protein complex with posaconazole (31)
or oteseconazole (49) has similar binding interactions with 22 amino
acids, on which oteseconazole (49) has an important H-bond with the
His-377 from its trifluoroethoxyphenyl oxygen (Fig. 14). His377 is
highly conserved in different fungal pathogens and also is specific for
fungi and not humans. This shows the importance of H-bonding in the
fungal CYP51 active site complex with inhibitors for potent and selec­
tive drug design. Key binding pocket interactions for fluconazole (24) in
LBP of S. cerevisiae and oteseconazole (49) in C. albicans CYP51 active
site are presented in Fig. 14 [130,134].
Despite these similarities in the CYP51 amino acid sequences and
Fig. 12. Some of important fluconazole hybrid antifungal agents.
M. Shafiei, et al. Bioorganic Chemistry 104 (2020) 104240
14
interactions of different inhibitors with them, allowing for development
of selective inhibitors with a wide spectrum of action in CYP51 human
pathogens, it is necessary to study the conformation and binding in­
teractions of each new inhibitor. This will permit and allow investiga­
tion of target-driven information in the context of target-based drug
discovery, setting new interactions on selective and non-conserved re­
sidues to reduce the risk of resistance in future [136,137].
Significant progress has been made with regard to information on
these X-ray crystal structures in characterization of CYP51 for anti­
fungal drug discovery. Several complex structures of CYP51 with in­
hibitors are available from RCSB Protein Data Bank (PDB) (rcsb.org).
The information on CYP51 structures is presented in Table 5
[127,134,136,138–142].
2.2.1. Structure activity relationship (SAR)
After the voriconazole (30) discovery, there were > 1000 flucona­
zole-derived structures that were synthesized and experimentally tested
at Pfizer, an inefficient and costly process [143]. Therefore, to design
and survey new azoles, as a leading class of important antifungal drugs,
a good model of structure–activity relationships could be cost and time
beneficial. From many studies on azole structure–activity relationships
and others surveying interactions in target protein with x-ray crystal­
lography or efficient docking methods, several useful pieces of in­
formation have been obtained. Here, we gathered, modeled and sum­
marized important structural features for future studies in the field.
In general, all azole drugs have a similar structural model (Fig. 15),
consisting of a Heme binding group, a three-atom linker, side-chain A
and side-chain B that are fully described below.
- Heme binding group:
This ring is crucial for the antifungal activity of the azole group
without any substitutions that bind to the linker with N-1 atom. The
basic N-3 of the imidazole ring or N-4 of the triazole or tetrazole ring is
positioned vertically to the porphyrin plane, as the iron of the active
site with the length of the coordinated bond of 1.2–2.1 A⁰. The basicity
of heteroaromatic nitrogen-containing rings does not have any re­
levance with the inhibitory potency of the azole structure; however, it is
directly associated with liver toxicity and drug-drug interactions. We
can also define basicity as iron-binding affinity. Thus, tetrazole with the
lowest basicity (-1.1 vs 2.3 for triazole and 6.8 for imidazole) has
minimum inhibitory for CYP450 and related adverse effects
[99,128,144].
- The three-atoms linker:
Although we named this linker as a three-atom linker, it could be
varied between 1 and 4 atoms. This three-atom shape makes a special
distance between different arms of structure, leading to greater potency
[145]. The atom number 1 of the linker group in azole structures is
commonly carbon and does not have any substitutions except topical
agents such as clotrimazole analogs and vinyl imidazole derivations.
For the atom number 2 and 3 in the linker, which are mostly a carbon,
chirality is important for antifungal activity [146,147]. Presence of an
oxygen atom as a hydroxyl group on C-2 has several beneficial effects
such as enhanced potency due to an indirect H-bond via water mole­
cules with active site, improved pharmacokinetics properties and water
Fig. 13. A. Sterol biosynthesis from Acetyl-CoA to ergosterol in fungi. B. The active site of CYP51[PDB: 4LXJ]: lanosrterol (green), heme (cyan), iron (orange),
oxygen(red) and Reaction steps catalyzed by C14-demethylase. (For interpretation of the references to colour in this figure legend, the reader is referred to the web
version of this article.)
M. Shafiei, et al. Bioorganic Chemistry 104 (2020) 104240
15
solubility, better tolerated and stable metabolism. Moreover, it is
needed to design prodrugs such as phosphate ester of fluconazole (24),
voriconazole (30) and ravuconazole (34) [44,148,149]. A methyl group
on C-3 could enhance potency against moulds. The chiral form gives
special conformation that side-chain B moves in antiperiplanar to side-
chain A and also this methyl group fills the binding pocket of C-13 of
lanosterol [46,150]. Other substitutions such as CH (double bond with
C-3) and two methyl or two F (non-chiral C-3) have also the desired
activity. In some of the first generation structures, C-3 became an
oxygen, sulfur or NR2 (oxime and hydrazine) (miconazole (2) and its
derivations), which maintains antifungal activity and also conforma­
tional restriction of structure with a ring on C-2 and C-3, such as
dioxalene (itraconazole (26) or ketoconazole (23)) or tetrahydrofuran
(posaconazole (31)); however, they do not enhance potency, or the
other pharmacokinetic properties and other groups such as ester,
amide, and carbamate are not appropriate for this replacement in an­
tifungal activity, in contrast to antitrypanosome cruzi and anti­
tubercular activity [151,152].
- Side chain A:
Side chain A in the structure that is always a halogenated phenyl, is
essential for inhibitory effect and appears to be located in the same
hydrophobic tunnel occupied with the 17-alkyl chain of lanosterol. It
Fig. 14. A: 3D structure of S. cerevisiae CYP51 complexed with fluconazole in the active site (PBD: 4WMZ). B: 3D structure of C. albicans CYP51 complexed with
oteseconazole in the active site (PBD: 5TZ1). Cyan dash line: metal coordination, Red dashes lines: water-mediated hydrogen bonding, green dash line: pi-stacking
interaction.
M. Shafiei, et al. Bioorganic Chemistry 104 (2020) 104240
16
has pi-pi stacking interactions with several residues like Y132 (a con­
served amino acid among fungus). Substitutions of the phenyl ring in all
carbons are tolerated except in groups larger than chlorine in C-2 and C-
6 that have unfavorable steric clashes and cause a decrease in the
binding affinity of inhibitors in the active site. A greater potency is
obtained from fluorine atoms in C-2 and C-4. However, other large
bulky groups such as 4-fluorophenyl could be replaced in para position
and maintain the CYP51 inhibition activity of molecules [145,153].
- Side chain B:
Side chain B area is similar to that of the heme-binding group as we
could design potent and favorable pharmacokinetic properties only
with optimization in this side chain. Many medicinal chemistry studies
survey various substitutions in this side chain as can be seen in third-
generation and hybrid structures (Fig. 7 and Fig. 12) [112].
In general, a linker binds the side chain B to C-3 of tree atom linker,
and antifungal activity differs from several different types of linkers
here. Studies evaluating this area show that ether, thioether, carbon
(–CH2-, -CHR-, -CR2-), -NR2, -SO2-, heteroaromatic or hetroaliphatic
(e.g., piperazine, piperidine) rings are well-tolerated and are good types
of linkage, while amide, ester, and -NHR reduce antifungal activity
[154–156]. Substitutions binding to the structure via the mentioned
linkers could make several hydrophobic, steric and important H-bond
interactions in the deep binding cleft of target protein. Before any de­
scription of this area, we divided azole drug into two groups based on
the size of the side chain, short-tail and long-tail structures.
For short tail structures, the linker may or may not have small
substitutions, like fluconazole (24), ravuconazole (34), and vor­
iconazole (30) structures. Small hydrophobic, electron-rich and elec­
tron-withdrawing groups (e.g., halogens, –CN, and halogenated het­
eroaromatic rings) enhance potency in the ortho and para-position of a
phenyl or heteroaromatic ring attached to the linker, with probable
steric and hydrophobic interactions (both aromatic ring and its sub­
stitution). In the voriconazole (30) case, fluorine on the pyrimidine ring
and Tyr122 of A. fumigatus form an H-bond interaction. Meta-position
substitutions are not favorable for activity [128,155].
For long-tail structures like itraconazole (26), posaconazole (31)
and VT-1598 (50), the para-position of a phenyl ring (like those for
short tail) could have a week H-bond acceptor substitution, preferably
oxygen (ether, carbonyl) and Nitrogen (NR2) that seems to form an H-
bond with some residue in the active site such as S-378 or H-374 and H-
377 (in the case of VT-1598 and oteseconazole) [147,155,156], and
also this H-bond acceptor group binds to another bulky aromatic group
with possible steric and van de waals interactions in the active site. In
the case of itraconazole-like drugs, a triazole-3-on group could enhance
pharmacokinetics of a compound. This area in the active site has en­
ough space for further modification of structures to improve potency,
physicochemical and pharmacokinetic of desired antifungal com­
pounds.
3. Concluding remarks and future directions
The number of hosts for invasive fungal infections is growing fast
due to an increase in circumstances that cause disturbed immune
function. In addition, the resistant rate of infective fungal spp to drugs
has improved with several known mechanisms like alteration in active
site residue by point mutations in ERG11 as the responsible gen for
CYP51, as well as ERG11 overexpression in fungal cells and MDR1
overexpression of efflux pump in fungi that may be concluded from
insufficient drug therapy. Thus, common drugs are ineffective and in­
adequate to deal with this situation [157–160]. Although, four gen­
erations of azoles as most diverse antifungal drugs are well developed
so far, the clinic has limited options for treatment. The options should
contain a wide spectrum action, parenteral or oral antifungal drug with
minimum drug-drug interaction and side-effects, effect on resistant
fungal infections, and acceptable pharmacokinetics properties. This
would never be achieved unless there are numerous studies by
Table 5
fungal CYP51, Drug − Target Complexes in PDB Databank (rcsb.org) as
Assessed in august 2019.
Organism Ligand PDB code Resolution (A⁰)
A.famigatus VNI 6CR2 2.38
A.famigatus VT-1598 5FRB 2.99
C. Albicans Itraconazole 5V5Z 2.9
C. Albicans Posaconazole 5FSA 2.86
C. Albicans VT-1161 5TZ1 2.0
C. Glabrata Itraconazole 5JLC 2.4
S. Cerevisiae Posaconazole 6E8Q 2.2
S. Cerevisiae Fluconazole 4WMZ 2.05
S. Cerevisiae VT-1161 5UL0 2.2
S. Cerevisiae Voriconazole 5HS1 2.1
S. Cerevisiae Lanosterol 4LXJ 1.9
S. Cerevisiae Itraconazole 5EQB 2.19
S. Cerevisiae (Y140F mutant) Itraconazole 4ZDY 2.02
S. Cerevisiae (Y140F mutant) Fluconazole 4ZDZ 2.3
S. Cerevisiae (Y140F mutant) Voriconazole 4ZE0 2.2
S. Cerevisiae (6464S mutant) Fluconazole 5ESY 2.15
S. Cerevisiae (G464S mutant) Itraconazole 5ESK 2.24
S. Cerevisiae (G73E mutant) Fluconazole 5ESF 2.25
S. Cerevisiae (G73E mutant) Itraconazole 5ESG 1.98
S. Cerevisiae (G73R mutant) Fluconazole 5ESE 2.2
S. Cerevisiae (G73W mutant) Itraconazole 5ESH 2.15
S. Cerevisiae (T3221 mutant) Itraconazole 5ESL 2.35
S. Cerevisiae (T3221 mutant) Fluconazole 5ESM 2.0
S. Cerevisiae (Y140Fmutant) Posaconazole 4ZE1 2.05
S. Cerevisiae (Y140H mutant) Itraconazole 4ZE2 2.3
S. Cerevisiae (Y140H mutant) Fluconazole 4ZE3 2.2
Fig. 15. Azole common structural model.
M. Shafiei, et al. Bioorganic Chemistry 104 (2020) 104240
17
medicinal chemistry scientist, the path started with fluconazole (24)
discovery and is on road with the newest 4th generation VT-1598 (50)
with improved characteristics. Albeit, in this way, azole is not the only
solution to the problem and identification of new targets are important
for efficient therapy. In this review, efforts were made to raise medic­
inal chemists’ awareness toward azole from their ancestor chlormida­
zole (1) to the most recent drug candidates in clinical trials and for the
first time introduce them in four generations with different new
structures. Moreover, hybrid antifungal agents as possible future gen­
eration which attracted high interest, were presented. The structure
activity relationship according to valid and authentic information from
SAR studies were also reported. Even though, in each study on the novel
efficient structures the target-based information, based on new X-ray of
CYP51 of wild or resistant types are unavoidable, we inform researchers
of the recently uploaded information on the protein data bank (PDB,
rcsb.org) for discovery and development of new members in the azole
family.
Declaration of Competing Interest
The authors declared that there is no conflict of interest.
Acknowledgements
This work was supported by a grant from the research council of
Tehran University of Medical Sciences (Grant no: 98-02-33-42922).
Consent for publication
Not applicable.
References
[1] G.D. Brown, D.W. Denning, N.A.R. Gow, S.M. Levitz, M.G. Netea, T.C. White,
Supplementary Materials for Hidden Killers : Human Fungal Infections 13 (2012)
4–6, https://doi.org/10.1126/scitranslmed.3004404.
[2] M.K. Kathiravan, A.B. Salake, A.S. Chothe, P.B. Dudhe, R.P. Watode, M.S. Mukta,
S. Gadhwe, The biology and chemistry of antifungal agents: A review, Bioorganic
Med. Chem. 20 (2012) 5678–5698, https://doi.org/10.1016/j.bmc.2012.04.045.
[3] N. Thamban Chandrika, E.K. Dennis, S.K. Shrestha, H.X. Ngo, K.D. Green, S.
Kwiatkowski, A.G. Deaciuc, L.P. Dwoskin, D.S. Watt, S. Garneau-Tsodikova, N,N′-
diaryl-bishydrazones in a biphenyl platform: Broad spectrum antifungal agents,
Eur. J. Med. Chem. (2019) 273–281. doi:10.1016/j.ejmech.2018.12.042.
[4] J.R. Perfect, The antifungal pipeline: A reality check, Nat. Rev. Drug Discov. 16
(2017) 603–616, https://doi.org/10.1038/nrd.2017.46.
[5] M. Ikeh, Y. Ahmed, J. Quinn, Phosphate Acquisition and Virulence in Human
Fungal Pathogens, Microorganisms. 5 (2017) 48, https://doi.org/10.3390/
microorganisms5030048.
[6] G.D. Brown, D.W. Denning, S.M. Levitz, Tackling human fungal infections, Science
(80-.). 336 (2012) 647. doi:10.1126/science.1222236.
[7] T. Roemer, D.J. Krysan, Unmet Clinical Needs, and New Approaches, Cold Spring
Harb Perspect Med. (2014) 1–14, https://doi.org/10.1101/cshperspect.a019703.
[8] J. Perlroth, B. Choi, B. Spellberg, Nosocomial fungal infections: Epidemiology,
diagnosis, and treatment, Med. Mycol. 45 (2007) 321–346, https://doi.org/10.
1080/13693780701218689.
[9] R. Calderone, N. Sun, F. Gay-Andrieu, W. Groutas, P. Weerawarna, S. Prasad,
D. Alex, D. Li, Antifungal drug discovery: The process and outcomes, Future
Microbiol. 9 (2014) 791–805, https://doi.org/10.2217/fmb.14.32.
[10] M.E. Rodrigues, S. Silva, J. Azeredo, M. Henriques, Novel strategies to fight
Candida species infection, Crit. Rev. Microbiol. 42 (2016) 594–606, https://doi.
org/10.3109/1040841X.2014.974500.
[11] M.A. Pfaller, D.J. Diekema, Epidemiology of invasive candidiasis: A persistent
public health problem, Clin. Microbiol. Rev. 20 (2007) 133–163, https://doi.org/
10.1128/CMR.00029-06.
[12] V. Onnis, A. De Logu, M.T. Cocco, R. Fadda, R. Meleddu, C. Congiu, 2-
Acylhydrazino-5-arylpyrrole derivatives: Synthesis and antifungal activity eva­
luation, Eur. J. Med. Chem. 44 (2009) 1288–1295, https://doi.org/10.1016/j.
ejmech.2008.08.003.
[13] A. Wald, W. Leisenring, J. van Burik, R.A. Bowden, Epidemiology of Aspergillus
Infections in a Large Cohort of Patients Undergoing Bone Marrow Transplantation,
J. Infect. Dis. 175 (1997) 1459–1466, https://doi.org/10.1086/516480.
[14] D.A. Enoch, H. Yang, S.H. Aliyu, C. Micallef, Human Fungal Pathogen
Identification (2017), https://doi.org/10.1007/978-1-4939-6515-1.
[15] R.A. Fromtling, Overview of medically important antifungal azole derivatives,
Clin. Microbiol. Rev. 1 (1988) 187–217, https://doi.org/10.1128/CMR.1.2.187.
[16] J.P. Utz, Chemotherapy for the Systemic Mycoses: The Prelude to Ketoconazole,
Clin. Infect. Dis. 2 (2011) 625–632, https://doi.org/10.1093/clinids/2.4.625.
[17] A. Butts, D.J. Krysan, Antifungal Drug Discovery: Something Old and Something
New, PLoS Pathog. 8 (2012) 9–11, https://doi.org/10.1371/journal.ppat.
1002870.
[18] A.H. Aly, A. Debbab, P. Proksch, Fifty years of drug discovery from fungi, Fungal
Divers. 50 (2011) 3–19, https://doi.org/10.1007/s13225-011-0116-y.
[19] T.K. Mazu, B.A. Bricker, H. Flores-Rozas, S.Y. Ablordeppey, The Mechanistic
Targets of Antifungal Agents: An Overview (2016), https://doi.org/10.2174/
1389557516666160118112103.
[20] D.S. Wishart, Y.D. Feunang, A.C. Guo, E.J. Lo, A. Marcu, J.R. Grant, T. Sajed, D.
Johnson, C. Li, Z. Sayeeda, N. Assempour, I. Iynkkaran, Y. Liu, A. MacIejewski, N.
Gale, A. Wilson, L. Chin, R. Cummings, Di. Le, A. Pon, C. Knox, M. Wilson,
DrugBank 5.0: A major update to the DrugBank database for 2018, Nucleic Acids
Res. 46 (2018) D1074–D1082. doi:10.1093/nar/gkx1037.
[21] H. Su, L. Han, X. Huang, Potential targets for the development of new antifungal
drugs, J. Antibiot. (Tokyo) 71 (2018) 978–991, https://doi.org/10.1038/s41429-
018-0100-9.
[22] N. Liu, J. Tu, G. Dong, Y. Wang, C. Sheng, Emerging New Targets for the
Treatment of Resistant Fungal Infections, J. Med. Chem. 61 (2018) 5484–5511,
https://doi.org/10.1021/acs.jmedchem.7b01413.
[23] C. Sheng, W. Zhang, New Lead Structures in Antifungal Drug Discovery, Curr.
Med. Chem. 18 (2011) 733–766, https://doi.org/10.2174/092986711794480113.
[24] L.R. Peyton, S. Gallagher, M. Hashemzadeh, Triazole antifungals: A review, Drugs
of Today. 51 (2015) 705–718, https://doi.org/10.1358/dot.2015.51.12.2421058.
[25] M.A. Hossain, M.A. Ghannoum, New investigational antifungal agents for treating
invasive fungal infections, Expert Opin. Invest. Drugs 9 (2005) 1797–1813,
https://doi.org/10.1517/13543784.9.8.1797.
[26] M.C.M. Marı́a Victoria Castelli, Estefanı́a Butassi, F.V.& S.A.Z. Laura A Svetaz,
Novel antifungal agents: a patent review (2011 - present), Expert Opin. Ther. Pat.
27 (2014) 415–426. doi:10.1517/13543776.2014.876993.
[27] J.A. Paiva, J.M. Pereira, New antifungal antibiotics, Curr. Opin. Infect. Dis. 26
(2013) 168–174, https://doi.org/10.1097/QCO.0b013e32835ebcb7.
[28] D.W. Woolley, Some biological effects produced by benzimidazole and their re­
versal by purines, J. Biol. Chem. (1944) 225–233.
[29] H. van den Bossche, Biochemical effects of miconazole on fungi—I, Biochem.
Pharmacol. 23 (2002) 887–899, https://doi.org/10.1016/0006-2952(74)90220-2.
[30] V.H.P.R. Seeliger, Pilzhemmende Wirkung eines neuen Benzimidazol-Derivates,
Mycoses 1 (1958) 162–171, https://doi.org/10.1111/j.1439-0507.1958.
tb04016.x.
[31] J.A. Maertens, History of the development of azole derivatives, Clin. Microbiol.
Infect. 10 (2004) 1–10, https://doi.org/10.1111/j.1470-9465.2004.00841.x.
[32] J.Y. Choi, L.M. Podust, W.R. Roush, Drug strategies targeting CYP51 in neglected
tropical diseases, Chem. Rev. 114 (2014) 11242–11271, https://doi.org/10.1021/
cr5003134.
[33] D. Allen, D. Wilson, R. Drew, J. Perfect, Azole antifungals: 35 years of invasive
fungal infection management, Expert Rev. Anti. Infect. Ther. 13 (2015) 787–798,
https://doi.org/10.1586/14787210.2015.1032939.
[34] V. Fainstein, G.P. Bodey, Cardiorespiratory Toxicity Due to Miconazole, Ann.
Intern. Med. 93 (2013) 432, https://doi.org/10.7326/0003-4819-93-3-432.
[35] P.R. Sawyer, R.N. Brogden, T.M. Speight, G.S. Avery, clotrimazole: A Review of its
Antifungal Activity and Therapeutic Efficacy, Drugs. 9 (1975) 424–447, https://
doi.org/10.2165/00003495-197509060-00003.
[36] D. Khanna, S. Bharti, Luliconazole for the treatment of fungal infections: an evi­
dence-based review, Core Evid. (2014) 113, https://doi.org/10.2147/ce.s49629.
[37] J. Heeres, L. Backx, J. Mostmans, J. Van Cutsem, Antimycotic imidazoles. Part 4.
Synthesis and antifungal activity of ketoconazole, a new potent orally active
broad-spectrum antifungal agent, J. Med. Chem. 22 (1979) 1003–1005.
[38] E. Drouhet, B. Dupont, Chronic Mucocutaneous Candidosis and Other Superficial
and Systemic Mycoses Successfully Treated with Ketoconazole, Clin. Infect. Dis. 2
(2011) 606–619, https://doi.org/10.1093/clinids/2.4.606.
[39] nizoral fda, (n.d.). www.fda.gov/Drugs/DrugSafety/ucm362415.htm.
[40] K. Richardson, The discovery and profile of fluconazole, J. Chemother. 2 (1990)
51–54, https://doi.org/10.1080/1120009X.1990.11738981.
[41] R. Cha, J.D. Sobel, Fluconazole for the treatment of candidiasis: 15 years experi­
ence, Expert Rev. Anti. Infect. Ther. 2 (2004) 357–366, https://doi.org/10.1586/
14787210.2.3.357.
[42] P.F. Troke, R.J. Andrews, G.W. Pye, K. Richardson, Fluconazole and other azoles:
Translation of in vitro activity to in vivo and clinical efficacy, Rev. Infect. Dis. 12
(1990) S276–S280, https://doi.org/10.1093/clinids/12.Supplement_3.S276.
[43] R.O. Darouiche, D. Abi-Said, D.P. Kontoyiannis, L.O. Gentry, E.J. Anaissie,
T. Williams, G.P. Bodey, J. Mera, C.L. Karl, O. Uzun, Management of Invasive
Candidal Infections: Results of a Prospective, Randomized, Multicenter Study of
Fluconazole Versus Amphotericin B and Review of the Literature, Clin. Infect. Dis.
23 (2011) 964–972, https://doi.org/10.1093/clinids/23.5.964.
[44] A. Bentley, M. Butters, S.P. Green, W.J. Learmonth, J.A. MacRae, M.C. Morland,
G. O’Connor, J. Skuse, The discovery and process development of a commercial
route to the water soluble prodrug, fosfluconazole, Org. Process Res. Dev. 6 (2002)
109–112, https://doi.org/10.1021/op010064+.
[45] S. Sobue, K. Tan, G. Layton, M. Eve, J.B. Sanderson, Pharmacokinetics of fos­
fluconazole and fluconazole following multiple intravenous administration of
fosfluconazole in healthy male volunteers, Br. J. Clin. Pharmacol. 58 (2004)
20–25, https://doi.org/10.1111/j.1365-2125.2004.02107.x.
[46] D. Abraham, Chemotherapeutic Agents, in: Burger’s Med. Chem. Drug Discov.,
John Sons, Inc, 2003: pp. 881–910.
[47] E.J. Corey, M. Chaykovsky, Dimethylsulfonium Methylide, A Reagent for Selective
M. Shafiei, et al. Bioorganic Chemistry 104 (2020) 104240
18
Oxirane Synthesis from Aldehydes and Ketones, J. Am. Chem. Soc. 84 (1962)
3782–3783, https://doi.org/10.1021/ja00878a046.
[48] E.J. Corey, M. Chaykovsky, Methylsulfinyl Carbanion (CH 3 -SO-CH 2). Formation
and Applications to Organic Synthesis, J. Am. Chem. Soc. 87 (1965) 1345–1353,
https://doi.org/10.1021/ja01084a033.
[49] E.J. Corey, M. Chaykovsky, Formation and Photochemical Rearrangement of β’-
Ketosulfoxonium Ylides, J. Am. Chem. Soc. 86 (1964) 1640–1641, https://doi.
org/10.1021/ja01062a040.
[50] J.J. Li, Corey-Chaykovsky epoxidation, Name React. (2002), https://doi.org/10.
1007/978-3-662-04835-1_64 77 77.
[51] M.J. Dolton, A.J. McLachlan, Optimizing azole antifungal therapy in the prophy­
laxis and treatment of fungal infections, Curr. Opin. Infect. Dis. 27 (2014)
493–500, https://doi.org/10.1097/QCO.0000000000000103.
[52] M.W. Pound, M.L. Townsend, V. Dimond, D. Wilson, R.H. Drew, Overview of
treatment options for invasive fungal infections, Med. Mycol. 49 (2011) 561–580,
https://doi.org/10.3109/13693786.2011.560197.
[53] C. Lass-Flörl, Triazole antifungal agents in invasive fungal infections: A com­
parative review, Drugs. 71 (2011) 2405–2419, https://doi.org/10.2165/
11596540-000000000-00000.
[54] S.W. Chapman, W.E. Dismukes, L.A. Proia, R.W. Bradsher, P.G. Pappas, M.G.
Threlkeld, C.A. Kauffman, Clinical Practice Guidelines for the Management of
Blastomycosis: 2008 Update by the Infectious Diseases Society of America, Clin.
Infect. Dis. 46 (2008) 1801–1812. doi:10.1086/588300.
[55] D.A. Stevens, J.Y. Lee, Analysis Of Compassionate Use Itraconazole Therapy For
Invasive Aspergillosis By The Niaid Mycoses Study Group Criteria, Arch. Intern.
Med. 157 (1997) 1857–1862.
[56] L. Willems, R. Van der Geest, K. de Beule, Itraconazole oral solution and in­
travenous formulations: a review of pharmacokinetics and pharmacodynamics, J.
Clin. Pharm. Ther. 26 (2001) 159–169 https://www.patientslikeme.com.
[57] T.K. Daneshmend, D.W. Warnock, Clinical Pharmacokinetics of Ketoconazole,
Clin. Pharmacokinet. 14 (1988) 13–34, https://doi.org/10.2165/00003088-
198814010-00002.
[58] L. Jeu, F.J. Piacenti, A.G. Lyakhovetskiy, H.B. Fung, New bugs Voriconazole, New
York. (2003).
[59] F. Ezzet, D. Wexler, R. Courtney, G. Krishna, J. Lim, M. Laughlin, Oral
Bioavailability of Posaconazole in Fasted Healthy Subjects, Clin. Pharmacokinet.
44 (2005) 211–220, https://doi.org/10.2165/00003088-200544020-00006.
[60] R. Bellmann, P. Smuszkiewicz, Pharmacokinetics of antifungal drugs: practical
implications for optimized treatment of patients, Infection 45 (2017) 737–779,
https://doi.org/10.1007/s15010-017-1042-z.
[61] S.R. Lipner, R.K. Scher, Efinaconazole in the treatment of onychomycosis, Infect.
Drug Resist. 8 (2015) 163–172, https://doi.org/10.2147/IDR.S69596.
[62] A.C. Pasqualotto, K.O. Thiele, L.Z. Goldani, Novel triazole antifungal drugs: focus
on isavuconazole, ravuconazole and albaconazole, Curr. Opin. Invest. Drugs 11
(2010) 165–174 http://www.ncbi.nlm.nih.gov/pubmed/20112166.
[63] A.J. Dietz, J.C. Barnard, K. van Rossem, A randomized, double-blind, multiple-
dose, placebo-controlled, dose escalation study with a 3-cohort parallel group
design to investigate the tolerability and pharmacokinetics of albaconazole in
healthy subjects, Clin. Pharmacol. Drug Dev. 3 (2014) 25–33, https://doi.org/10.
1002/cpdd.72.
[64] C. Lin, H. Kim, E. Radwanski, M. Affrime, M. Brannan, M.N. Cayen,
Pharmacokinetics and metabolism of genaconazole, a potent antifungal drug, in
men, Antimicrob. Agents Chemother. 40 (1996) 92–96, https://doi.org/10.1128/
AAC.40.1.92.
[65] N. Sun, Y. Xie, C. Sheng, Y. Cao, W. Zhang, H. Chen, G. Fan, In vivo pharmaco­
kinetics and in vitro antifungal activity of iodiconazole, a new triazole, determined
by microdialysis sampling, Int. J. Antimicrob. Agents 41 (2013) 229–235, https://
doi.org/10.1016/j.ijantimicag.2012.10.020.
[66] Y. Kamai, M. Kubota, T. Fukuoka, Y. Kamai, N. Maeda, T. Hosokawa,
T. Shibayama, K. Uchida, H. Yamaguchi, S. Kuwahara, Efficacy of CS-758, a novel
triazole, against experimental fluconazole-resistant oropharyngeal candidiasis in
mice, Antimicrob. Agents Chemother. 47 (2003) 601–606, https://doi.org/10.
1128/AAC.47.2.601-606.2003.
[67] C. Girmenia, E. Finolezzi, New-generation triazole antifungal drugs: review of the
Phase II and III trials, Clin. Investig. (Lond) 1 (2011) 1577–1594, https://doi.org/
10.4155/cli.11.137.
[68] L.B. Johnson, C.A. Kuffman, Voriconazole: A new triazole antifungal agent, Rev.
ANTI-INFECTIVE AGENTS. 36 (2003) 630 637 doi:1058-4838/2003/3605-
0014$15.00.
[69] F. Sabatelli, R. Patel, P.A. Mann, C.A. Mendrick, C.C. Norris, R. Hare,
D. Loebenberg, T.A. Black, P.M. McNicholas, In vitro activities of posaconazole,
fluconazole, itraconazole, voriconazole, and amphotericin B against a large col­
lection of clinically important molds and yeasts, Antimicrob. Agents Chemother.
50 (2006) 2009–2015, https://doi.org/10.1128/AAC.00163-06.
[70] F. Bennett, A.K. Saksena, R.G. Lovey, Y.T. Liu, N.M. Patel, P. Pinto, R. Pike, E. Jao,
V.M. Girijavallabhan, A.K. Ganguly, D. Loebenberg, H. Wang, A. Cacciapuoti,
E. Moss, F. Menzel, R.S. Hare, A. Nomeir, Hydroxylated analogues of the orally
active broad spectrum antifungal, Sch 51048 (1), and the discovery of posaco­
nazole [Sch 56592; 2 or (S, S)-5], Bioorganic Med. Chem. Lett. 16 (2006) 186–190,
https://doi.org/10.1016/j.bmcl.2005.09.031.
[71] A. Nomeir, B.N. Pramanik, L. Heimark, F. Bennett, J. Veals, P. Bartner, M. Hilbert,
A. Saksena, P. McNamara, Posaconazole (Noxafil, SCH 56592), a new azole an­
tifungal drug, was a discovery based on the isolation and mass spectral char­
acterization of a circulating metabolite of an earlier lead (SCH 51048), J. Mass
Spectrom. 43 (2008) 509–517, https://doi.org/10.1002/jms.1341.
[72] I.I. Raad, J.R. Graybill, A.B. Bustamante, O.A. Cornely, V. Gaona-Flores, C. Afif,
D.R. Graham, R.N. Greenberg, S. Hadley, A. Langston, R. Negroni, J.R. Perfect,
P. Pitisuttithum, A. Restrepo, G. Schiller, L. Pedicone, A.J. Ullmann, Safety of
Long-Term Oral Posaconazole Use in the Treatment of Refractory Invasive Fungal
Infections, Clin. Infect. Dis. 42 (2006) 1726–1734, https://doi.org/10.1086/
504328.
[73] O.A. Cornely, J. Maertens, D.J. Winston, J. Perfect, A.J. Ullmann, T.J. Walsh,
D. Helfgott, J. Holowiecki, D. Stockelberg, Y.-T. Goh, M. Petrini, C. Hardalo,
R. Suresh, D. Angulo-Gonzalez, Posaconazole vs. Fluconazole or Itraconazole
Prophylaxis in Patients with Neutropenia, N. Engl. J. Med. 356 (2007) 348–359,
https://doi.org/10.1056/nejmoa061094.
[74] M. Andrew J. Ullmann, M.D., Jeffrey H. Lipton, M.D., David H. Vesole, M.D., Ph.
D., Pranatharthi Chandrasekar, M.D., Amelia Langston, M.D., Stefano R.
Tarantolo, M.D., Hildegard Greinix, M.D., Wellington Morais de Azevedo, M.D.,
Ph.D., Vijay Reddy, M.D., Navd, Posaconazole or Fluconazole for Prophylaxis in
Severe Graft-versus-Host Disease, N. Engl. J. Med. 356 (2007) 335–347.
[75] T.J. Walsh, I. Raad, T.F. Patterson, P. Chandrasekar, G.R. Donowitz, R. Graybill,
R.E. Greene, R. Hachem, S. Hadley, R. Herbrecht, A. Langston, A. Louie, P. Ribaud,
B.H. Segal, D.A. Stevens, J.H. van Burik, C.S. White, G. Corcoran, J. Gogate,
G. Krishna, L. Pedicone, C. Hardalo, J.R. Perfect, Treatment of Invasive
Aspergillosis with Posaconazole in Patients Who Are Refractory to or Intolerant of
Conventional Therapy: An Externally Controlled Trial, Clin. Infect. Dis. 44 (2007)
2–12, https://doi.org/10.1086/508774.
[76] T. Patel, S. Dhillon, Efinaconazole: First global approval, Drugs. 73 (2013)
1977–1983, https://doi.org/10.1007/s40265-013-0152-x.
[77] A. Schmitt-hoffmann, B. Roos, M. Heep, M. Schleimer, E. Weidekamm, T. Brown,
M. Roehrle, C. Beglinger, Single-Ascending-Dose Pharmacokinetics and Safety of
the Novel Broad-Spectrum Antifungal Triazole BAL4815 after Intravenous
Infusions (50, 100, and 200 Milligrams) and Oral Administrations (100, 200, and
400 Milligrams) of Its Prodrug, BAL8557, in Healthy, Antimicrob. Agents
Chemother. 50 (2006) 279–285, https://doi.org/10.1128/AAC.50.1.279-285.
2006.
[78] boyd 83, (n.d.). https://www.aspergillus.org.uk/content/bal-4815bal-8557-anti­
fungal-agent.
[79] S. Seyedmousavi, P.E. Verweij, J.W. Mouton, Isavuconazole, a broad-spectrum
triazole for the treatment of systemic fungal diseases, Expert Rev. Anti. Infect.
Ther. 13 (2015) 9–27, https://doi.org/10.1586/14787210.2015.990382.
[80] N.N. Pettit, P.L. Carver, Isavuconazole: A New Option for the Management of
Invasive Fungal Infections, Ann. Pharmacother. 49 (2015) 825–842, https://doi.
org/10.1177/1060028015581679.
[81] H. Yamaguchi, Potential of Ravuconazole and its Prodrugs as the New
OralTherapeutics for Onychomycosis, Med. Mycol. J. 57 (2016) E93–E110,
https://doi.org/10.3314/mmj.16-00006.
[82] J. Capilla, M. Ortoneda, F.J. Pastor, J. Guarro, In vitro antifungal activities of the
new triazole UR-9825 against clinically important filamentous fungi, Antimicrob.
Agents Chemother. 45 (2001) 2635–2637, https://doi.org/10.1128/AAC.45.9.
2635-2637.2001.
[83] clinical trial, (n.d.). https://clinicaltrials.gov.
[84] Y.N. Cheng, Z.H. Jiang, L.S. Sun, Z.Y. Su, M.M. Zhang, H.L. Li, Synthesis of 1, 2, 4-
triazole benzoyl arylamine derivatives and their high antifungal activities, Eur. J.
Med. Chem. 200 (2020) 112463, https://doi.org/10.1016/j.ejmech.2020.112463.
[85] Z. Ding, T. Ni, F. Xie, Y. Hao, S. Yu, X. Chai, Y. Jin, T. Wang, Y. Jiang, D. Zhang,
Design, synthesis, and structure-activity relationship studies of novel triazole
agents with strong antifungal activity against Aspergillus fumigatus, Bioorganic
Med. Chem. Lett. 30 (2020) 126951, https://doi.org/10.1016/j.bmcl.2020.
126951.
[86] F. Xie, T. Ni, Z. Ding, Y. Hao, R. Wang, R. Wang, T. Wang, X. Chai, S. Yu, Y. Jin,
Y. Jiang, D. Zhang, Design, synthesis, and in vitro evaluation of novel triazole
analogues featuring isoxazole moieties as antifungal agents, Bioorg. Chem. 101
(2020) 103982, https://doi.org/10.1016/j.bioorg.2020.103982.
[87] İ.S. Doğan, S. Saraç, S. Sari, D. Kart, Ş. Eşsiz Gökhan, İ. Vural, S. Dalkara, New
azole derivatives showing antimicrobial effects and their mechanism of antifungal
activity by molecular modeling studies, Eur. J. Med. Chem. 130 (2017) 124–138,
https://doi.org/10.1016/j.ejmech.2017.02.035.
[88] V.W. Rabelo, T.F. Santos, L. Terra, M.V. Santana, H.C. Castro, C.R. Rodrigues,
P.A. Abreu, Targeting CYP51 for drug design by the contributions of molecular
modeling, Fundam. Clin. Pharmacol. 31 (2017) 37–53, https://doi.org/10.1111/
fcp.12230.
[89] Y. Zhang, V.K.R. Tangadanchu, R.R.Y. Bheemanaboina, Y. Cheng, C.H. Zhou,
Novel carbazole-triazole conjugates as DNA-targeting membrane active potentia­
tors against clinical isolated fungi, Eur. J. Med. Chem. 155 (2018) 579–589,
https://doi.org/10.1016/j.ejmech.2018.06.022.
[90] Z. Jiang, J. Gu, C. Wang, S. Wang, N. Liu, Y. Jiang, G. Dong, Y. Wang, Y. Liu,
J. Yao, Z. Miao, W. Zhang, C. Sheng, Design, synthesis and antifungal activity of
novel triazole derivatives containing substituted 1,2,3-triazole-piperdine side
chains, Eur. J. Med. Chem. 82 (2014) 490–497, https://doi.org/10.1016/j.ejmech.
2014.05.079.
[91] S.K. Shrestha, A. Garzan, S. Garneau-Tsodikova, Novel alkylated azoles as potent
antifungals, Eur. J. Med. Chem. 133 (2017) 309–318, https://doi.org/10.1016/j.
ejmech.2017.03.075.
[92] A. Qian, Y. Zheng, R. Wang, J. Wei, Y. Cui, X. Cao, Y. Yang, Design, synthesis, and
structure-activity relationship studies of novel tetrazole antifungal agents with
potent activity, broad antifungal spectrum and high selectivity, Bioorganic Med.
Chem. Lett. 28 (2018) 344–350, https://doi.org/10.1016/j.bmcl.2017.12.040.
[93] P.D. Patel, M.R. Patel, B. Kocsis, E. Kocsis, S.M. Graham, A.R. Warren,
S.M. Nicholson, B. Billack, F.R. Fronczek, T.T. Talele, Design, synthesis and de­
termination of antifungal activity of 5(6)-substituted benzotriazoles, Eur. J. Med.
M. Shafiei, et al. Bioorganic Chemistry 104 (2020) 104240
19
Chem. 45 (2010) 2214–2222, https://doi.org/10.1016/j.ejmech.2010.01.062.
[94] W. Wang, S. Wang, Y. Liu, G. Dong, Y. Cao, Z. Miao, J. Yao, W. Zhang, C. Sheng,
Novel conformationally restricted triazole derivatives with potent antifungal ac­
tivity, Eur. J. Med. Chem. 45 (2010) 6020–6026, https://doi.org/10.1016/j.
ejmech.2010.09.070.
[95] Y. Zhang, G.L.V. Damu, S.F. Cui, J.L. Mi, V.K.R. Tangadanchu, C.H. Zhou,
Discovery of potential antifungal triazoles: Design, synthesis, biological evalua­
tion, and preliminary antifungal mechanism exploration, Medchemcomm. 8
(2017) 1631–1639, https://doi.org/10.1039/c7md00112f.
[96] H. Tang, J. Wu, W. Zhang, L. Zhao, Y.H. Zhang, C.W. Shen, Design, synthesis and
biological evaluation of novel non-azole derivatives as potential antifungal agents,
Chinese Chem. Lett. 26 (2015) 1161–1164, https://doi.org/10.1016/j.cclet.2015.
04.030.
[97] N.P. Wiederhold, L.K. Najvar, E.P. Garvey, S.R. Brand, X. Xu, E.A. Ottinger,
A. Alimardanov, J. Cradock, M. Behnke, W.J. Hoekstra, R.J. Schotzinger,
R. Jaramillo, M. Olivo, W.R. Kirkpatrick, T.F. Patterson, The fungal cyp51 in­
hibitor vt-1129 is efficacious in an experimental model of cryptococcal meningitis,
Antimicrob. Agents Chemother. 62 (2018) 1–10, https://doi.org/10.1128/AAC.
01071-1.
[98] S.R. Brand, T.P. Degenhardt, K. Person, J.D. Sobel, P. Nyirjesy, R.J. Schotzinger,
A. Tavakkol, A phase 2, randomized, double-blind, placebo-controlled, dose-ran­
ging study to evaluate the efficacy and safety of orally administered VT-1161 in
the treatment of recurrent vulvovaginal candidiasis, Am. J. Obstet. Gynecol. 624
(e1–624) (2018) e9, https://doi.org/10.1016/j.ajog.2018.03.001.
[99] W.J. Hoekstra, E.P. Garvey, W.R. Moore, S.W. Rafferty, C.M. Yates,
R.J. Schotzinger, Design and optimization of highly-selective fungal CYP51 in­
hibitors, Bioorganic Med. Chem. Lett. 24 (2014) 3455–3458, https://doi.org/10.
1016/j.bmcl.2014.05.068.
[100] C.M. Yates, E.P. Garvey, S.R. Shaver, R.J. Schotzinger, W.J. Hoekstra, Design and
optimization of highly-selective, broad spectrum fungal CYP51 inhibitors,
Bioorganic Med. Chem. Lett. 27 (2017) 3243–3248, https://doi.org/10.1016/j.
bmcl.2017.06.037.
[101] N.P. Wiederhold, H.P. Patterson, B.H. Tran, C.M. Yates, R.J. Schotzinger,
E.P. Garvey, Fungal-specific Cyp51 inhibitor VT-1598 demonstrates in vitro ac­
tivity against Candida and Cryptococcus species, endemic fungi, including
Coccidioides species, Aspergillus species and Rhizopus arrhizus, J. Antimicrob.
Chemother. 73 (2018) 404–408, https://doi.org/10.1093/jac/dkx410.
[102] A.G.S. Warrilow, J.E. Parker, C.L. Price, W.D. Nes, E.P. Garvey, W.J. Hoekstra,
R.J. Schotzinger, D.E. Kelly, S.L. Kelly, The investigational drug VT-1129 is a
highly potent inhibitor of Cryptococcus species CYP51 but only weakly inhibits the
human enzyme, Antimicrob. Agents Chemother. 60 (2016) 4530–4538, https://
doi.org/10.1128/AAC.00349-16.
[103] S. Brand, T. Degenhardt, P. Nyirjesy, J. Sobel, C. Handelsman, K. Person,
R. Schotzinger, A. Tavakkol, 15: To evaluate the efficacy and safety of VT-1161, a
potent, highly selective inhibitor of fungal CYP51, in treating women with a
documented history of recurrent vulvovaginal candidiasis (RVVC), Am. J. Obstet.
Gynecol. 215 (2016) S821, https://doi.org/10.1016/j.ajog.2016.09.016.
[104] S.Q. Wang, Y.F. Wang, Z. Xu, Tetrazole hybrids and their antifungal activities, Eur.
J. Med. Chem. (2019) 225–234, https://doi.org/10.1016/j.ejmech.2019.03.023.
[105] N.P. Wiederhold, The antifungal arsenal: alternative drugs and future targets, Int.
J. Antimicrob. Agents 51 (2018) 333–339, https://doi.org/10.1016/j.ijantimicag.
2017.09.002.
[106] N.P. Wiederhold, L.K. Najvar, R. Jaramillo, M. Olivo, G. Catano, T.F. Patterson,
L.F. Shubitz, H.T. Trinh, C.M. Yates, R.J. Schotzinger, E.P. Garvey, The novel
fungal Cyp51 inhibitor VT-1598 is efficacious in experimental models of central
nervous system coccidioidomycosis caused by Coccidioides posadasii and
Coccidioides immitis, Antimicrob. Agents Chemother. 62 (2018) 1–7, https://doi.
org/10.1128/AAC.02258-17.
[107] N.P. Wiederhold, S.R. Lockhart, L.K. Najvar, E.L. Berkow, R. Jaramillo, M. Olivo,
E.P. Garvey, C.M. Yates, R.J. Schotzinger, G. Catano, T.F. Patterson, The Fungal
Cyp51-Specific Inhibitor VT-1598 Demonstrates In Vitro and In Vivo Activity
against Candida auris, Antimicrob. Agents Chemother. (2019), https://doi.org/10.
1128/AAC.02233-18.
[108] B. Meunier, Hybrid Molecules with a Dual Mode of Action : Dream or Reality ? †,
Acc. Chem. Res. 41 (2008) 69–77.
[109] F. Terms, An overview of molecular hybrids in drug discovery, Expert Opin. Drug
Discov. (2016), https://doi.org/10.1517/17460441.2016.1135125.
[110] H.R. Martins, J.D. De Souza, F. Marcelo, P.B. Victor, H.O.M. Jarbas, R.M. Verly,
Glycotriazole - peptides derived from the peptide HSP1: synergistic effect of
triazole and saccharide rings on the antifungal activity, Amino Acids 49 (2017)
1389–1400, https://doi.org/10.1007/s00726-017-2441-2.
[111] S. Wu, Y. Zhang, X. He, X. Che, S. Wang, Y. Liu, Y. Jiang, N. Liu, G. Dong, J. Yao,
Z. Miao, Y. Wang, From Antidiabetic to Antifungal : Discovery of Highly Potent
Triazole – Thiazolidinedione Hybrids as Novel Antifungal Agents, ChemMedChem
9 (2014) 2639–2646, https://doi.org/10.1002/cmdc.201402320.
[112] S. Emami, E. Ghobadi, S. Saednia, S.M. Hashemi, Current advances of triazole
alcohols derived from fluconazole: Design, in vitro and in silico studies, Eur. J.
Med. Chem. (2019) 173–194, https://doi.org/10.1016/j.ejmech.2019.03.020.
[113] N. Belskaya, J. Subbotina, S. Lesogorova, Synthesis of 2 H -1, 2, 3-Triazoles, Top
Heterocycl Chem. 1 (2015) 51–116, https://doi.org/10.1007/7081.
[114] Y. Wang, K. Xu, G. Bai, L. Huang, Q. Wu, W. Pan, S. Yu, Synthesis and Antifungal
Activity of Novel Triazole Compounds Containing Piperazine Moiety, Molecules
19 (2014) 11333–11340, https://doi.org/10.3390/molecules190811333.
[115] N.D. Thanh, D.S. Hai, V.T. Ngoc Bich, P.T. Thu Hien, N.T. Ky Duyen, N.T. Mai, T.T.
Dung, V.N. Toan, H.T. Kim Van, L.H. Dang, D.N. Toan, T.T. Thanh Van, Efficient
click chemistry towards novel 1H-1,2,3-triazole-tethered 4H-chromene−D-
glucose conjugates: Design, synthesis and evaluation of in vitro antibacterial,
MRSA and antifungal activities, Eur. J. Med. Chem. (2019) 454–471. doi:10.1016/
j.ejmech.2019.01.060.
[116] Y. Zou, Q. Zhao, J. Liao, H. Hu, S. Yu, X. Chai, M. Xu, Q. Wu, Bioorganic &
Medicinal Chemistry Letters New triazole derivatives as antifungal agents :
Synthesis via click reaction, in vitro evaluation and molecular docking studies,
Bioorg. Med. Chem. Lett. 22 (2012) 2959–2962, https://doi.org/10.1016/j.bmcl.
2012.02.042.
[117] N. Fu, S. Wang, Y. Zhang, C. Zhang, D. Yang, L. Weng, B. Zhao, L. Wang, Efficient
click chemistry towards fatty acids containing 1,2,3-triazole: Design and synthesis
as potential antifungal drugs for Candida albicans, Eur. J. Med. Chem. (2017),
https://doi.org/10.1016/j.ejmech.2017.05.001.
[118] V.S. Pore, N.G. Aher, P.K. Shukla, Design and synthesis of fluconazole / bile acid
conjugate using click reaction, Tetrahedron 62 (2006) 11178–11186, https://doi.
org/10.1016/j.tet.2006.09.021.
[119] K. Bozorov, J. Zhao, H.A. Aisa, 1,2,3-Triazole-containing hybrids as leads in
medicinal chemistry: A recent overview, Bioorganic Med. Chem. 27 (2019)
3511–3531, https://doi.org/10.1016/j.bmc.2019.07.005.
[120] A. Gluszynska, Biological potential of carbazole derivatives, Eur. J. Med. Chem. 94
(2015) 405–426, https://doi.org/10.1016/j.ejmech.2015.02.059.
[121] R. Elias, R.I. Benhamou, Q.Z. Jaber, O. Dorot, S.L. Zada, K. Oved, E. Pichinuk,
M. Fridman, Antifungal activity, mode of action variability, and subcellular dis­
tribution of coumarin-based antifungal azoles, Eur. J. Med. Chem. 179 (2019)
779–790, https://doi.org/10.1016/j.ejmech.2019.07.003.
[122] G. Han, N. Liu, C. Li, J. Tu, Z. Li, C. Sheng, Discovery of Novel Fungal Lanosterol
14 α -Demethylase (CYP51)/ Histone Deacetylase (HDAC) Dual Inhibitors to treat
azole-resistant candidiasis, J. Med. Chem. Discov. Apr (2020). doi:10.1021/acs.
jmedchem.0c00102.
[123] X. Fang, D. Li, V. Kumar, R. Tangadanchu, L. Gopala, W. Gao, Novel potentially
antifungal hybrids of 5-flucytosine and fluconazole : Design, synthesis and
bioactive evaluation, Bioorg. Med. Chem. Lett. (2017), https://doi.org/10.1016/j.
bmcl.2017.10.020.
[124] T. Režen, N. Debeljak, D. Kordiš, D. Rozman, New aspects on lanosterol 14α-de­
methylase and cytochrome P450 evolution: Lanosterol/cycloartenol diversifica­
tion and lateral transfer, J. Mol. Evol. 59 (2004) 51–58, https://doi.org/10.1007/
s00239-004-2603-1.
[125] E. Desmond, S. Gribaldo, Phylogenomics of Sterol Synthesis: Insights into the
Origin, Evolution, and Diversity of a Key Eukaryotic Feature, Genome Biol. Evol. 1
(2009) 364–381, https://doi.org/10.1093/gbe/evp036.
[126] M.V. Keniya, M. Sabherwal, R.K. Wilson, M.A. Woods, A.A. Sagatova,
J.D.A. Tyndall, Crystal Structures of Full-Length Lanosterol 14α-Demethylases of
Prominent Fungal Pathogens Candida albicans and Candida glabrata Provide Tools
for Antifungal Discovery, Antimicrob. Agents Chemother. 62 (2018)
e01134–e1218.
[127] F. Hannemann, A. Bichet, K.M. Ewen, R. Bernhardt, Cytochrome P450 systems-
biological variations of electron transport chains, Biochim. Biophys. Acta - Gen.
Subj. 1770 (2007) 330–344, https://doi.org/10.1016/j.bbagen.2006.07.017.
[128] T.Y. Hargrove, Z. Wawrzak, D.C. Lamb, F.P. Guengerich, G.I. Lepesheva,
Structure-functional characterization of cytochrome P450 Sterol 14α-Demethylase
(CYP51B) from aspergillus fumigatus and molecular basis for the development of
antifungal drugs, J. Biol. Chem. 290 (2015) 23916–23934, https://doi.org/10.
1074/jbc.M115.677310.
[129] B.C. Monk, A.A. Sagatova, P. Hosseini, Y.N. Ruma, R.K. Wilson, M.V. Keniya,
Fungal Lanosterol 14α-demethylase: A target for next-generation antifungal de­
sign, Biochim. Biophys. Acta - Proteins Proteomics. (2019), https://doi.org/10.
1016/j.bbapap.2019.02.008.
[130] J. Zhang, L. Li, Q. Lv, L. Yan, Y. Wang, Y. Jiang, The fungal CYP51s: Their func­
tions, structures, related drug resistance, and inhibitors, Front. Microbiol. 10
(2019), https://doi.org/10.3389/fmicb.2019.00691.
[131] F.V. Galina. Lepesheva, Tatiana Y. Hargrove1, Girish Rachakonda, Zdzislaw
Wawrzak, Sébastien Pomel, Sandrine Cojean, Pius N. Nde, W. David Nes, Charles
W. Locuson, M. Wade Calcutt, Michael R. Waterman, J. Scott Daniels, Philippe M.
Loiseau, VFV as a new effective CYP51 structure-derived drug candidate for
Chagas disease and visceral leishmaniasis, J. Infect. Dis. April 15 (2015) 1–25.
doi:10.1093/infdis/jiv228.
[132] F. Villalta, M.C. Dobish, P.N. Nde, Y.Y. Kleshchenko, T.Y. Hargrove, C.A. Johnson,
M.R. Waterman, J.N. Johnston, G.I. Lepesheva, VNI cures acute and chronic ex­
perimental chagas disease, J. Infect. Dis. 208 (2013) 504–511, https://doi.org/10.
1093/infdis/jit042.
[133] G.I. Lepesheva, H.W. Park, T.Y. Hargrove, B. Vanhollebeke, Z. Wawrzak,
J.M. Harp, M. Sundaramoorthy, W.D. Nes, E. Pays, M. Chaudhuri, F. Villalta,
M.R. Waterman, Crystal structures of Trypanosoma brucei sterol 14α-demethylase
and implications for selective treatment of human infections, J. Biol. Chem. 285
(2010) 1773–1780, https://doi.org/10.1074/jbc.M109.067470.
[134] T.Y. Hargrove, L. Friggeri, Z. Wawrzak, A. Qi, W.J. Hoekstra, R.J. Schotzinger,
J.D. York, F. Peter Guengerich, G.I. Lepesheva, Structural analyses of Candida
albicans sterol 14α-demethylase complexed with azole drugs address the mole­
cular basis of azole-mediated inhibition of fungal sterol biosynthesis, J. Biol.
Chem. 292 (2017) 6728–6743, https://doi.org/10.1074/jbc.M117.778308.
[135] G.I. Lepesheva, M.R. Waterman, Structural basis for conservation in the CYP51
family, Biochim. Biophys. Acta - Proteins Proteomics. 2011 (1814) 88–93, https://
doi.org/10.1016/j.bbapap.2010.06.006.
[136] A.A. Sagatova, M.V. Keniya, R.K. Wilson, B.C. Monk, J.D.A. Tyndall, Structural
insights into binding of the antifungal drug fluconazole to Saccharomyces cere­
visiae lanosterol 14α-demethylase, Antimicrob. Agents Chemother. 59 (2015)
4982–4989, https://doi.org/10.1128/AAC.00925-15.
M. Shafiei, et al. Bioorganic Chemistry 104 (2020) 104240
20
[137] C. Sheng, S. Chen, H. Ji, G. Dong, X. Che, W. Wang, Z. Miao, J. Yao, J. Lü, W. Guo,
W. Zhang, Evolutionary trace analysis of CYP51 family: Implication for site-di­
rected mutagenesis and novel antifungal drug design, J. Mol. Model. 16 (2010)
279–284, https://doi.org/10.1007/s00894-009-0527-9.
[138] B.C. Monk, M.V. Keniya, M. Sabherwal, R.K. Wilson, D.O. Graham, H.F. Hassan,
D. Chen, J.D.A. Tyndall, Azole resistance reduces susceptibility to the tetrazole
antifungal VT-1161, Antimicrob. Agents Chemother. 63 (2019) 1–19, https://doi.
org/10.1128/AAC.02114-18.
[139] 5ul0 116, (n.d.). https://www.rcsb.org/structure/5ul0.
[140] B.C. Monk, T.M. Tomasiak, M.V. Keniya, F.U. Huschmann, J.D.A. Tyndall,
J.D. O’Connell, R.D. Cannon, J.G. McDonald, A. Rodriguez, J.S. Finer-Moore,
R.M. Stroud, Architecture of a single membrane spanning cytochrome P450 sug­
gests constraints that orient the catalytic domain relative to a bilayer, Proc. Natl.
Acad. Sci. 111 (2014) 3865–3870, https://doi.org/10.1073/pnas.1324245111.
[141] L. Friggeri, T.Y. Hargrove, Z. Wawrzak, A.L. Blobaum, G. Rachakonda,
C.W. Lindsley, F. Villalta, W.D. Nes, M. Botta, F.P. Guengerich, G.I. Lepesheva,
Sterol 14α-Demethylase Structure-Based Design of VNI ((R)- N-(1-(2,4-
Dichlorophenyl)-2-(1 H-imidazol-1-yl)ethyl)-4-(5-phenyl-1,3,4-oxadiazol-2-yl)
benzamide)) Derivatives to Target Fungal Infections: Synthesis, Biological
Evaluation, and Crystallographic A, J. Med. Chem. 61 (2018) 5679–5691, https://
doi.org/10.1021/acs.jmedchem.8b00641.
[142] T.Y. Hargrove, E.P. Garvey, W.J. Hoekstra, C.M. Yates, Z. Wawrzak,
G. Rachakonda, F. Villalta, G.I. Lepesheva, Crystal structure of the new in­
vestigational drug candidate VT-1598 in complex with Aspergillus fumigatus
sterol 14α-demethylase provides insights into its broad-spectrum Antifungal
Activity, Antimicrob. Agents Chemother. 61 (2017), https://doi.org/10.1128/
AAC.00570-17.
[143] D.W. Denning, M.J. Bromley, How to bolster the antifungal pipeline, Science (80-.
). 347 (2015) 1414–1416. doi:10.1126/science.aaa6097.
[144] M.A. Correia, P.R. Ortiz de Montellano, Inhibition of Cytochrome P450 Enzymes,
Cytochrome P450 Struct, Mech. Biochem. (2005) 247–322, https://doi.org/10.
2460/ajvr.72.11.1505.
[145] H. Ji, W. Zhang, Y. Zhou, M. Zhang, J. Zhu, Y. Song, J. Lü, J. Zhu, A three-di­
mensional model of lanosterol 14α-demethylase of Candida albicans and its in­
teraction with azole antifungals, J. Med. Chem. 43 (2000) 2493–2505, https://doi.
org/10.1021/jm990589g.
[146] K. Tasaka, A.; Kitazaki, T.; Tsuchimori, N.; Matsushita, Y.; Hayashi, R.; Okonogi, K.
; Itoh, Optically active antifungal azoles. VII. Synthesis and antifungal activity of
stereoisomers of 2-[(1R,2R)-2-(2,4-difluorophenyl)-2-hydroxy-1-methyl-3-(1H-1,
2,4-triazol- 1-yl)propyl]-4-[4-(2,2,3,3-tetrafluoropropoxy)phenyl]-3(2H,4H)- 1,2,
4-triazolone (TAK-, Chem. Pharm. Bull. 45 (1997) 321–326. doi:10.1248/cpb.45.
321.
[147] D.M. Rotstein, D.J. Kertesz, K.A.M. Walker, D.C. Swinney, Stereoisomers of
Ketoconazole: Preparation and Biological Activity, J. Med. Chem. 35 (1992)
2818–2825, https://doi.org/10.1021/jm00093a015.
[148] J. Bartroli, E. Turmo, M. Alguero, E. Boncompte, M.L. Vericat, L. Conte, J. Ramis,
M. Merlos, J. Garcia-Rafanell, J. Forn, ChemInform Abstract: New Azole
Antifungals. Part 2. Synthesis and Antifungal Activity of Heterocyclecarboxamide
Derivatives of 3-Amino-2-aryl-1-azolyl-2-butanol, ChemInform. 29 (2010) no-no,
https://doi.org/10.1002/chin.199836174.
[149] G. Klopman, D. Ptchelintsev, Antifungal triazole alcohols: A comparative analysis
of structure-activity, structure-teratogenicity and structure-therapeutic index re­
lationships using the Multiple Computer-Automated Structure Evaluation (Multi-
CASE) methodology, J. Comput. Aided Mol. Des. 7 (1993) 349–362, https://doi.
org/10.1007/BF00125508.
[150] J. Bartroli, E. Turmo, M. Algueró, E. Boncompte, M.L. Vericat, J. GARCIA-
RAFANELL, J. FORN, Synthesis and antifungal activity of new azole derivatives
containing an N-acylmorpholine ring, J. Med. Chem. 38 (1995) 3918–3932.
doi:10.1021/jm00020a005.
[151] D. De Vita, F. Pandolfi, R. Cirilli, L. Scipione, R. Di Santo, L. Friggeri, M. Mori,
D. Fiorucci, G. Maccari, R.S.A. Christopher, C. Zamperini, V. Pau, A. De Logu,
S. Tortorella, M. Botta, Discovery of in vitro antitubercular agents through in silico
ligand-based approaches, Eur. J. Med. Chem. 121 (2016) 169–180, https://doi.
org/10.1016/j.ejmech.2016.05.032.
[152] L. Friggeri, T.Y. Hargrove, G. Rachakonda, A.D. Williams, Z. Wawrzak, R. Di
Santo, D. De Vita, M.R. Waterman, S. Tortorella, F. Villalta, G.I. Lepesheva,
Structural basis for rational design of inhibitors targeting Trypanosoma cruzi
Sterol 14α-demethylase: Two regions of the enzyme molecule potentiate its in­
hibition, J. Med. Chem. 57 (2014) 6704–6717, https://doi.org/10.1021/
jm500739f.
[153] L.M. Podust, T.L. Poulos, M.R. Waterman, Crystal structure of cytochrome P450 14
-sterol demethylase (CYP51) from Mycobacterium tuberculosis in complex with
azole inhibitors, Proc. Natl. Acad. Sci. 98 (2002) 3068–3073, https://doi.org/10.
1073/pnas.061562898.
[154] F. Chevreuil, A. Landreau, D. Seraphin, G. Larcher, J.P. Bouchara, P. Richomme,
Synthesis and antifungal activity of new thienyl and aryl conazoles, J. Enzyme
Inhib. Med. Chem. 21 (2006) 293–303, https://doi.org/10.1080/
14756360600700640.
[155] C. Sheng, W. Zhang, H. Ji, M. Zhang, Y. Song, H. Xu, J. Zhu, Z. Miao, Q. Jiang,
J. Yao, Y. Zhou, J. Zhu, J. Lü, Structure-based optimization of azole antifungal
agents by CoMFA, CoMSIA, and molecular docking, J. Med. Chem. 49 (2006)
2512–2525, https://doi.org/10.1021/jm051211n.
[156] J. Bartroli, E. Turmo, M. Algueró, E. Boncompte, M.L. Vericat, L. Conte, J. Ramis,
M. Merlos, New Azole Antifungals. 3. Synthesis and Antifungal Activity of 3-
Substituted-4(3H)-quinazolinones, J. Med. Chem. 41 (1998) 1869–1882.
[157] N.M. Revie, K.R. Iyer, N. Robbins, L.E. Cowen, Antifungal drug resistance: evo­
lution, mechanisms and impact, Curr. Opin. Microbiol. 45 (2018) 70–76, https://
doi.org/10.1016/j.mib.2018.02.005.
[158] T.P. Salci, M. Negri, A.K.R. Abadio, T.I.E. Svidzinski, É.S. Kioshima, Targeting
Candida spp. to develop antifungal agents, Drug Discov. Today. 23 (2018)
802–814, https://doi.org/10.1016/j.drudis.2018.01.003.
[159] A.M. Fuentefria, B. Pippi, D.F. Dalla Lana, K.K. Donato, S.F. de Andrade,
Antifungals discovery: an insight into new strategies to combat antifungal re­
sistance, Lett. Appl. Microbiol. 66 (2018) 2–13, https://doi.org/10.1111/lam.
12820.
[160] J.A. Parente-Rocha, A.M. Bailão, A.C. Amaral, C.P. Taborda, J.D. Paccez,
C.L. Borges, M. Pereira, Antifungal Resistance, Metabolic Routes as Drug Targets,
and New Antifungal Agents: An Overview about Endemic Dimorphic Fungi,
Mediators Inflamm. 2017 (2017), https://doi.org/10.1155/2017/9870679.
M. Shafiei, et al. Bioorganic Chemistry 104 (2020) 104240
21

More Related Content

What's hot

Promising Sudanese Medicinal Plants with Antibacterial Activity - a Review Ar...
Promising Sudanese Medicinal Plants with Antibacterial Activity - a Review Ar...Promising Sudanese Medicinal Plants with Antibacterial Activity - a Review Ar...
Promising Sudanese Medicinal Plants with Antibacterial Activity - a Review Ar...Dheeraj Vasu
 
Evidencias2006i 100619025249-phpapp02
Evidencias2006i 100619025249-phpapp02Evidencias2006i 100619025249-phpapp02
Evidencias2006i 100619025249-phpapp02UNICA-Publicidad
 
Multidrug Resistance Pattern of Staphylococcus Aureus Isolates in Maiduguri M...
Multidrug Resistance Pattern of Staphylococcus Aureus Isolates in Maiduguri M...Multidrug Resistance Pattern of Staphylococcus Aureus Isolates in Maiduguri M...
Multidrug Resistance Pattern of Staphylococcus Aureus Isolates in Maiduguri M...Scientific Review
 
Choudhary2020
Choudhary2020Choudhary2020
Choudhary2020Babahgaru
 
The skin microbiome a focus on pathogens and their
The skin microbiome a focus on pathogens and theirThe skin microbiome a focus on pathogens and their
The skin microbiome a focus on pathogens and theirSantiagoLopez411547
 
Role of Nonfermenting Gram-Negative Bacilli Responsible for Respiratory Tract...
Role of Nonfermenting Gram-Negative Bacilli Responsible for Respiratory Tract...Role of Nonfermenting Gram-Negative Bacilli Responsible for Respiratory Tract...
Role of Nonfermenting Gram-Negative Bacilli Responsible for Respiratory Tract...Premier Publishers
 
The efficacy of some medicinal plants used locally within Transmara west, Nar...
The efficacy of some medicinal plants used locally within Transmara west, Nar...The efficacy of some medicinal plants used locally within Transmara west, Nar...
The efficacy of some medicinal plants used locally within Transmara west, Nar...IOSRJPBS
 
Evaluation of calvatia craniformis mushroom activity in
Evaluation  of calvatia craniformis mushroom activity in Evaluation  of calvatia craniformis mushroom activity in
Evaluation of calvatia craniformis mushroom activity in Alexander Decker
 
A PROPOSAL ON BIOFILM FORMATION AND ANTIBIOTIC SUSCEPTIBILITY PATTERN OF Stap...
A PROPOSAL ON BIOFILM FORMATION AND ANTIBIOTIC SUSCEPTIBILITY PATTERN OF Stap...A PROPOSAL ON BIOFILM FORMATION AND ANTIBIOTIC SUSCEPTIBILITY PATTERN OF Stap...
A PROPOSAL ON BIOFILM FORMATION AND ANTIBIOTIC SUSCEPTIBILITY PATTERN OF Stap...Shrezjana Mainali
 
Reconstructing Exposures From The Uk Chemical Warfare Agent Human Research Pr...
Reconstructing Exposures From The Uk Chemical Warfare Agent Human Research Pr...Reconstructing Exposures From The Uk Chemical Warfare Agent Human Research Pr...
Reconstructing Exposures From The Uk Chemical Warfare Agent Human Research Pr...guest971b1073
 
Hamdy Eldoksh cv-recent
Hamdy Eldoksh cv-recent Hamdy Eldoksh cv-recent
Hamdy Eldoksh cv-recent Hamdy Eldoksh
 
PlP syllabus BSc General BURDWAN UNIBERSITY
PlP syllabus BSc General BURDWAN UNIBERSITYPlP syllabus BSc General BURDWAN UNIBERSITY
PlP syllabus BSc General BURDWAN UNIBERSITYarnab das
 
Akindele1442016BJPR30974-1
Akindele1442016BJPR30974-1Akindele1442016BJPR30974-1
Akindele1442016BJPR30974-1Peter Akindele
 

What's hot (17)

Promising Sudanese Medicinal Plants with Antibacterial Activity - a Review Ar...
Promising Sudanese Medicinal Plants with Antibacterial Activity - a Review Ar...Promising Sudanese Medicinal Plants with Antibacterial Activity - a Review Ar...
Promising Sudanese Medicinal Plants with Antibacterial Activity - a Review Ar...
 
Evidencias2006i 100619025249-phpapp02
Evidencias2006i 100619025249-phpapp02Evidencias2006i 100619025249-phpapp02
Evidencias2006i 100619025249-phpapp02
 
Cmdr 209 (1)
Cmdr 209 (1)Cmdr 209 (1)
Cmdr 209 (1)
 
Multidrug Resistance Pattern of Staphylococcus Aureus Isolates in Maiduguri M...
Multidrug Resistance Pattern of Staphylococcus Aureus Isolates in Maiduguri M...Multidrug Resistance Pattern of Staphylococcus Aureus Isolates in Maiduguri M...
Multidrug Resistance Pattern of Staphylococcus Aureus Isolates in Maiduguri M...
 
Choudhary2020
Choudhary2020Choudhary2020
Choudhary2020
 
The skin microbiome a focus on pathogens and their
The skin microbiome a focus on pathogens and theirThe skin microbiome a focus on pathogens and their
The skin microbiome a focus on pathogens and their
 
Role of Nonfermenting Gram-Negative Bacilli Responsible for Respiratory Tract...
Role of Nonfermenting Gram-Negative Bacilli Responsible for Respiratory Tract...Role of Nonfermenting Gram-Negative Bacilli Responsible for Respiratory Tract...
Role of Nonfermenting Gram-Negative Bacilli Responsible for Respiratory Tract...
 
Clinical distribution of biofilm forming staphylococcus aureus and its sensit...
Clinical distribution of biofilm forming staphylococcus aureus and its sensit...Clinical distribution of biofilm forming staphylococcus aureus and its sensit...
Clinical distribution of biofilm forming staphylococcus aureus and its sensit...
 
The efficacy of some medicinal plants used locally within Transmara west, Nar...
The efficacy of some medicinal plants used locally within Transmara west, Nar...The efficacy of some medicinal plants used locally within Transmara west, Nar...
The efficacy of some medicinal plants used locally within Transmara west, Nar...
 
Evaluation of calvatia craniformis mushroom activity in
Evaluation  of calvatia craniformis mushroom activity in Evaluation  of calvatia craniformis mushroom activity in
Evaluation of calvatia craniformis mushroom activity in
 
A PROPOSAL ON BIOFILM FORMATION AND ANTIBIOTIC SUSCEPTIBILITY PATTERN OF Stap...
A PROPOSAL ON BIOFILM FORMATION AND ANTIBIOTIC SUSCEPTIBILITY PATTERN OF Stap...A PROPOSAL ON BIOFILM FORMATION AND ANTIBIOTIC SUSCEPTIBILITY PATTERN OF Stap...
A PROPOSAL ON BIOFILM FORMATION AND ANTIBIOTIC SUSCEPTIBILITY PATTERN OF Stap...
 
C.musae review
C.musae reviewC.musae review
C.musae review
 
Reconstructing Exposures From The Uk Chemical Warfare Agent Human Research Pr...
Reconstructing Exposures From The Uk Chemical Warfare Agent Human Research Pr...Reconstructing Exposures From The Uk Chemical Warfare Agent Human Research Pr...
Reconstructing Exposures From The Uk Chemical Warfare Agent Human Research Pr...
 
Hamdy Eldoksh cv-recent
Hamdy Eldoksh cv-recent Hamdy Eldoksh cv-recent
Hamdy Eldoksh cv-recent
 
PlP syllabus BSc General BURDWAN UNIBERSITY
PlP syllabus BSc General BURDWAN UNIBERSITYPlP syllabus BSc General BURDWAN UNIBERSITY
PlP syllabus BSc General BURDWAN UNIBERSITY
 
Akindele1442016BJPR30974-1
Akindele1442016BJPR30974-1Akindele1442016BJPR30974-1
Akindele1442016BJPR30974-1
 
Antimicrobial therapy
Antimicrobial therapyAntimicrobial therapy
Antimicrobial therapy
 

Similar to Shafiei2020

A trends of salmonella and antibiotic resistance
A trends of salmonella and antibiotic resistanceA trends of salmonella and antibiotic resistance
A trends of salmonella and antibiotic resistanceAlexander Decker
 
A trends of salmonella and antibiotic resistance
A trends of salmonella and antibiotic resistanceA trends of salmonella and antibiotic resistance
A trends of salmonella and antibiotic resistanceAlexander Decker
 
The pharmacology and efficacy of antifungals: a literature review
The pharmacology and efficacy of antifungals: a literature reviewThe pharmacology and efficacy of antifungals: a literature review
The pharmacology and efficacy of antifungals: a literature reviewPARUL UNIVERSITY
 
THE PHARMACOLOGY AND EFFICACY OF ANTIFUNGALS: A LITERATURE REVIEW
THE PHARMACOLOGY AND EFFICACY OF ANTIFUNGALS: A LITERATURE REVIEWTHE PHARMACOLOGY AND EFFICACY OF ANTIFUNGALS: A LITERATURE REVIEW
THE PHARMACOLOGY AND EFFICACY OF ANTIFUNGALS: A LITERATURE REVIEWPARUL UNIVERSITY
 
pheromone traps for assessment and monitoring محطة بحوث وقاية النباتات 2019.pptx
pheromone traps for assessment and monitoring محطة بحوث وقاية النباتات 2019.pptxpheromone traps for assessment and monitoring محطة بحوث وقاية النباتات 2019.pptx
pheromone traps for assessment and monitoring محطة بحوث وقاية النباتات 2019.pptxAbdallah Albeltagy
 
An Overview of Chemotherapy of Tuberculosis: A Review
An Overview of Chemotherapy of Tuberculosis: A ReviewAn Overview of Chemotherapy of Tuberculosis: A Review
An Overview of Chemotherapy of Tuberculosis: A ReviewJing Zang
 
Analysis of used disinfectants and antiseptics correlated with the occurrence...
Analysis of used disinfectants and antiseptics correlated with the occurrence...Analysis of used disinfectants and antiseptics correlated with the occurrence...
Analysis of used disinfectants and antiseptics correlated with the occurrence...iosrphr_editor
 
Project amr by Arghya & Arnab
Project amr by Arghya & ArnabProject amr by Arghya & Arnab
Project amr by Arghya & ArnabArghya Chowdhury
 
ANTIBIOTICS (Tetracycline)
ANTIBIOTICS (Tetracycline)ANTIBIOTICS (Tetracycline)
ANTIBIOTICS (Tetracycline)Esam Yahya
 
Antibiotic resistant-genes-in-natural-environment-2168-9881.1000104
Antibiotic resistant-genes-in-natural-environment-2168-9881.1000104Antibiotic resistant-genes-in-natural-environment-2168-9881.1000104
Antibiotic resistant-genes-in-natural-environment-2168-9881.1000104science journals
 
Deciphering drug resistance in mtb using wgs
Deciphering drug resistance in mtb using wgsDeciphering drug resistance in mtb using wgs
Deciphering drug resistance in mtb using wgsVictorAkinseyeOluwat
 
Bio pesticides production
Bio pesticides productionBio pesticides production
Bio pesticides productionKrishna Annepu
 
Study of Bacterial Isolates and their Susceptibility Pattern in Chronic Suppu...
Study of Bacterial Isolates and their Susceptibility Pattern in Chronic Suppu...Study of Bacterial Isolates and their Susceptibility Pattern in Chronic Suppu...
Study of Bacterial Isolates and their Susceptibility Pattern in Chronic Suppu...iosrjce
 

Similar to Shafiei2020 (20)

A trends of salmonella and antibiotic resistance
A trends of salmonella and antibiotic resistanceA trends of salmonella and antibiotic resistance
A trends of salmonella and antibiotic resistance
 
A trends of salmonella and antibiotic resistance
A trends of salmonella and antibiotic resistanceA trends of salmonella and antibiotic resistance
A trends of salmonella and antibiotic resistance
 
The pharmacology and efficacy of antifungals: a literature review
The pharmacology and efficacy of antifungals: a literature reviewThe pharmacology and efficacy of antifungals: a literature review
The pharmacology and efficacy of antifungals: a literature review
 
THE PHARMACOLOGY AND EFFICACY OF ANTIFUNGALS: A LITERATURE REVIEW
THE PHARMACOLOGY AND EFFICACY OF ANTIFUNGALS: A LITERATURE REVIEWTHE PHARMACOLOGY AND EFFICACY OF ANTIFUNGALS: A LITERATURE REVIEW
THE PHARMACOLOGY AND EFFICACY OF ANTIFUNGALS: A LITERATURE REVIEW
 
Antimicrobial activity of Antibiotics and Antiseptics (Dettol and Betadine) a...
Antimicrobial activity of Antibiotics and Antiseptics (Dettol and Betadine) a...Antimicrobial activity of Antibiotics and Antiseptics (Dettol and Betadine) a...
Antimicrobial activity of Antibiotics and Antiseptics (Dettol and Betadine) a...
 
project mutamba
project mutambaproject mutamba
project mutamba
 
pheromone traps for assessment and monitoring محطة بحوث وقاية النباتات 2019.pptx
pheromone traps for assessment and monitoring محطة بحوث وقاية النباتات 2019.pptxpheromone traps for assessment and monitoring محطة بحوث وقاية النباتات 2019.pptx
pheromone traps for assessment and monitoring محطة بحوث وقاية النباتات 2019.pptx
 
An Overview of Chemotherapy of Tuberculosis: A Review
An Overview of Chemotherapy of Tuberculosis: A ReviewAn Overview of Chemotherapy of Tuberculosis: A Review
An Overview of Chemotherapy of Tuberculosis: A Review
 
Analysis of used disinfectants and antiseptics correlated with the occurrence...
Analysis of used disinfectants and antiseptics correlated with the occurrence...Analysis of used disinfectants and antiseptics correlated with the occurrence...
Analysis of used disinfectants and antiseptics correlated with the occurrence...
 
ODONTOLOGIA
ODONTOLOGIAODONTOLOGIA
ODONTOLOGIA
 
ODONTOLOGIA
ODONTOLOGIAODONTOLOGIA
ODONTOLOGIA
 
Project amr by Arghya & Arnab
Project amr by Arghya & ArnabProject amr by Arghya & Arnab
Project amr by Arghya & Arnab
 
ISAVUCONAZOL Review
ISAVUCONAZOL ReviewISAVUCONAZOL Review
ISAVUCONAZOL Review
 
ANTIBIOTICS (Tetracycline)
ANTIBIOTICS (Tetracycline)ANTIBIOTICS (Tetracycline)
ANTIBIOTICS (Tetracycline)
 
Antibiotic resistant-genes-in-natural-environment-2168-9881.1000104
Antibiotic resistant-genes-in-natural-environment-2168-9881.1000104Antibiotic resistant-genes-in-natural-environment-2168-9881.1000104
Antibiotic resistant-genes-in-natural-environment-2168-9881.1000104
 
Oxazolidina
OxazolidinaOxazolidina
Oxazolidina
 
Deciphering drug resistance in mtb using wgs
Deciphering drug resistance in mtb using wgsDeciphering drug resistance in mtb using wgs
Deciphering drug resistance in mtb using wgs
 
Tamizhazhagan.1
Tamizhazhagan.1Tamizhazhagan.1
Tamizhazhagan.1
 
Bio pesticides production
Bio pesticides productionBio pesticides production
Bio pesticides production
 
Study of Bacterial Isolates and their Susceptibility Pattern in Chronic Suppu...
Study of Bacterial Isolates and their Susceptibility Pattern in Chronic Suppu...Study of Bacterial Isolates and their Susceptibility Pattern in Chronic Suppu...
Study of Bacterial Isolates and their Susceptibility Pattern in Chronic Suppu...
 

Recently uploaded

SOLUBLE PATTERN RECOGNITION RECEPTORS.pptx
SOLUBLE PATTERN RECOGNITION RECEPTORS.pptxSOLUBLE PATTERN RECOGNITION RECEPTORS.pptx
SOLUBLE PATTERN RECOGNITION RECEPTORS.pptxkessiyaTpeter
 
Spermiogenesis or Spermateleosis or metamorphosis of spermatid
Spermiogenesis or Spermateleosis or metamorphosis of spermatidSpermiogenesis or Spermateleosis or metamorphosis of spermatid
Spermiogenesis or Spermateleosis or metamorphosis of spermatidSarthak Sekhar Mondal
 
Cultivation of KODO MILLET . made by Ghanshyam pptx
Cultivation of KODO MILLET . made by Ghanshyam pptxCultivation of KODO MILLET . made by Ghanshyam pptx
Cultivation of KODO MILLET . made by Ghanshyam pptxpradhanghanshyam7136
 
Natural Polymer Based Nanomaterials
Natural Polymer Based NanomaterialsNatural Polymer Based Nanomaterials
Natural Polymer Based NanomaterialsAArockiyaNisha
 
PossibleEoarcheanRecordsoftheGeomagneticFieldPreservedintheIsuaSupracrustalBe...
PossibleEoarcheanRecordsoftheGeomagneticFieldPreservedintheIsuaSupracrustalBe...PossibleEoarcheanRecordsoftheGeomagneticFieldPreservedintheIsuaSupracrustalBe...
PossibleEoarcheanRecordsoftheGeomagneticFieldPreservedintheIsuaSupracrustalBe...Sérgio Sacani
 
Biological Classification BioHack (3).pdf
Biological Classification BioHack (3).pdfBiological Classification BioHack (3).pdf
Biological Classification BioHack (3).pdfmuntazimhurra
 
Biopesticide (2).pptx .This slides helps to know the different types of biop...
Biopesticide (2).pptx  .This slides helps to know the different types of biop...Biopesticide (2).pptx  .This slides helps to know the different types of biop...
Biopesticide (2).pptx .This slides helps to know the different types of biop...RohitNehra6
 
Orientation, design and principles of polyhouse
Orientation, design and principles of polyhouseOrientation, design and principles of polyhouse
Orientation, design and principles of polyhousejana861314
 
Presentation Vikram Lander by Vedansh Gupta.pptx
Presentation Vikram Lander by Vedansh Gupta.pptxPresentation Vikram Lander by Vedansh Gupta.pptx
Presentation Vikram Lander by Vedansh Gupta.pptxgindu3009
 
CALL ON ➥8923113531 🔝Call Girls Kesar Bagh Lucknow best Night Fun service 🪡
CALL ON ➥8923113531 🔝Call Girls Kesar Bagh Lucknow best Night Fun service  🪡CALL ON ➥8923113531 🔝Call Girls Kesar Bagh Lucknow best Night Fun service  🪡
CALL ON ➥8923113531 🔝Call Girls Kesar Bagh Lucknow best Night Fun service 🪡anilsa9823
 
Unlocking the Potential: Deep dive into ocean of Ceramic Magnets.pptx
Unlocking  the Potential: Deep dive into ocean of Ceramic Magnets.pptxUnlocking  the Potential: Deep dive into ocean of Ceramic Magnets.pptx
Unlocking the Potential: Deep dive into ocean of Ceramic Magnets.pptxanandsmhk
 
Boyles law module in the grade 10 science
Boyles law module in the grade 10 scienceBoyles law module in the grade 10 science
Boyles law module in the grade 10 sciencefloriejanemacaya1
 
Lucknow 💋 Russian Call Girls Lucknow Finest Escorts Service 8923113531 Availa...
Lucknow 💋 Russian Call Girls Lucknow Finest Escorts Service 8923113531 Availa...Lucknow 💋 Russian Call Girls Lucknow Finest Escorts Service 8923113531 Availa...
Lucknow 💋 Russian Call Girls Lucknow Finest Escorts Service 8923113531 Availa...anilsa9823
 
Stunning ➥8448380779▻ Call Girls In Panchshil Enclave Delhi NCR
Stunning ➥8448380779▻ Call Girls In Panchshil Enclave Delhi NCRStunning ➥8448380779▻ Call Girls In Panchshil Enclave Delhi NCR
Stunning ➥8448380779▻ Call Girls In Panchshil Enclave Delhi NCRDelhi Call girls
 
Nightside clouds and disequilibrium chemistry on the hot Jupiter WASP-43b
Nightside clouds and disequilibrium chemistry on the hot Jupiter WASP-43bNightside clouds and disequilibrium chemistry on the hot Jupiter WASP-43b
Nightside clouds and disequilibrium chemistry on the hot Jupiter WASP-43bSérgio Sacani
 
Raman spectroscopy.pptx M Pharm, M Sc, Advanced Spectral Analysis
Raman spectroscopy.pptx M Pharm, M Sc, Advanced Spectral AnalysisRaman spectroscopy.pptx M Pharm, M Sc, Advanced Spectral Analysis
Raman spectroscopy.pptx M Pharm, M Sc, Advanced Spectral AnalysisDiwakar Mishra
 
G9 Science Q4- Week 1-2 Projectile Motion.ppt
G9 Science Q4- Week 1-2 Projectile Motion.pptG9 Science Q4- Week 1-2 Projectile Motion.ppt
G9 Science Q4- Week 1-2 Projectile Motion.pptMAESTRELLAMesa2
 

Recently uploaded (20)

SOLUBLE PATTERN RECOGNITION RECEPTORS.pptx
SOLUBLE PATTERN RECOGNITION RECEPTORS.pptxSOLUBLE PATTERN RECOGNITION RECEPTORS.pptx
SOLUBLE PATTERN RECOGNITION RECEPTORS.pptx
 
Spermiogenesis or Spermateleosis or metamorphosis of spermatid
Spermiogenesis or Spermateleosis or metamorphosis of spermatidSpermiogenesis or Spermateleosis or metamorphosis of spermatid
Spermiogenesis or Spermateleosis or metamorphosis of spermatid
 
Cultivation of KODO MILLET . made by Ghanshyam pptx
Cultivation of KODO MILLET . made by Ghanshyam pptxCultivation of KODO MILLET . made by Ghanshyam pptx
Cultivation of KODO MILLET . made by Ghanshyam pptx
 
Natural Polymer Based Nanomaterials
Natural Polymer Based NanomaterialsNatural Polymer Based Nanomaterials
Natural Polymer Based Nanomaterials
 
PossibleEoarcheanRecordsoftheGeomagneticFieldPreservedintheIsuaSupracrustalBe...
PossibleEoarcheanRecordsoftheGeomagneticFieldPreservedintheIsuaSupracrustalBe...PossibleEoarcheanRecordsoftheGeomagneticFieldPreservedintheIsuaSupracrustalBe...
PossibleEoarcheanRecordsoftheGeomagneticFieldPreservedintheIsuaSupracrustalBe...
 
Biological Classification BioHack (3).pdf
Biological Classification BioHack (3).pdfBiological Classification BioHack (3).pdf
Biological Classification BioHack (3).pdf
 
9953056974 Young Call Girls In Mahavir enclave Indian Quality Escort service
9953056974 Young Call Girls In Mahavir enclave Indian Quality Escort service9953056974 Young Call Girls In Mahavir enclave Indian Quality Escort service
9953056974 Young Call Girls In Mahavir enclave Indian Quality Escort service
 
Biopesticide (2).pptx .This slides helps to know the different types of biop...
Biopesticide (2).pptx  .This slides helps to know the different types of biop...Biopesticide (2).pptx  .This slides helps to know the different types of biop...
Biopesticide (2).pptx .This slides helps to know the different types of biop...
 
Orientation, design and principles of polyhouse
Orientation, design and principles of polyhouseOrientation, design and principles of polyhouse
Orientation, design and principles of polyhouse
 
Presentation Vikram Lander by Vedansh Gupta.pptx
Presentation Vikram Lander by Vedansh Gupta.pptxPresentation Vikram Lander by Vedansh Gupta.pptx
Presentation Vikram Lander by Vedansh Gupta.pptx
 
CALL ON ➥8923113531 🔝Call Girls Kesar Bagh Lucknow best Night Fun service 🪡
CALL ON ➥8923113531 🔝Call Girls Kesar Bagh Lucknow best Night Fun service  🪡CALL ON ➥8923113531 🔝Call Girls Kesar Bagh Lucknow best Night Fun service  🪡
CALL ON ➥8923113531 🔝Call Girls Kesar Bagh Lucknow best Night Fun service 🪡
 
Unlocking the Potential: Deep dive into ocean of Ceramic Magnets.pptx
Unlocking  the Potential: Deep dive into ocean of Ceramic Magnets.pptxUnlocking  the Potential: Deep dive into ocean of Ceramic Magnets.pptx
Unlocking the Potential: Deep dive into ocean of Ceramic Magnets.pptx
 
Boyles law module in the grade 10 science
Boyles law module in the grade 10 scienceBoyles law module in the grade 10 science
Boyles law module in the grade 10 science
 
Engler and Prantl system of classification in plant taxonomy
Engler and Prantl system of classification in plant taxonomyEngler and Prantl system of classification in plant taxonomy
Engler and Prantl system of classification in plant taxonomy
 
Lucknow 💋 Russian Call Girls Lucknow Finest Escorts Service 8923113531 Availa...
Lucknow 💋 Russian Call Girls Lucknow Finest Escorts Service 8923113531 Availa...Lucknow 💋 Russian Call Girls Lucknow Finest Escorts Service 8923113531 Availa...
Lucknow 💋 Russian Call Girls Lucknow Finest Escorts Service 8923113531 Availa...
 
CELL -Structural and Functional unit of life.pdf
CELL -Structural and Functional unit of life.pdfCELL -Structural and Functional unit of life.pdf
CELL -Structural and Functional unit of life.pdf
 
Stunning ➥8448380779▻ Call Girls In Panchshil Enclave Delhi NCR
Stunning ➥8448380779▻ Call Girls In Panchshil Enclave Delhi NCRStunning ➥8448380779▻ Call Girls In Panchshil Enclave Delhi NCR
Stunning ➥8448380779▻ Call Girls In Panchshil Enclave Delhi NCR
 
Nightside clouds and disequilibrium chemistry on the hot Jupiter WASP-43b
Nightside clouds and disequilibrium chemistry on the hot Jupiter WASP-43bNightside clouds and disequilibrium chemistry on the hot Jupiter WASP-43b
Nightside clouds and disequilibrium chemistry on the hot Jupiter WASP-43b
 
Raman spectroscopy.pptx M Pharm, M Sc, Advanced Spectral Analysis
Raman spectroscopy.pptx M Pharm, M Sc, Advanced Spectral AnalysisRaman spectroscopy.pptx M Pharm, M Sc, Advanced Spectral Analysis
Raman spectroscopy.pptx M Pharm, M Sc, Advanced Spectral Analysis
 
G9 Science Q4- Week 1-2 Projectile Motion.ppt
G9 Science Q4- Week 1-2 Projectile Motion.pptG9 Science Q4- Week 1-2 Projectile Motion.ppt
G9 Science Q4- Week 1-2 Projectile Motion.ppt
 

Shafiei2020

  • 1. Contents lists available at ScienceDirect Bioorganic Chemistry journal homepage: www.elsevier.com/locate/bioorg History of the development of antifungal azoles: A review on structures, SAR, and mechanism of action Mohammad Shafieia , Lee Peytonb , Mehrnoosh Hashemzadehc,⁎ , Alireza Foroumadia,⁎ a Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran b Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine and Science, Rochester, MN, USA c University of Arizona college of medicine Phoenix and Pima college, Tucson, AZ, USA A R T I C L E I N F O Keywords: Invasive fungal infection Lanosterol 14α-demethylase Azole Structure activity relationship Drug development Molecular hybrids A B S T R A C T With the increasing risk of invasive and life threating fungal infections, there is now a great concern regarding the lower discovery rate of antifungal drugs in comparison to antimicrobial agents. Drugs conventionally used in clinics are not adequate enough to combat the increasing fungal infections, especially fungal forms resistant to fluconazole. Among the limited antifungal agents in clinics, azoles have the largest number of drug candidates in clinical trials and are partly marketed due to the particular focus of pharmaceutical companies and medicinal scientific centers. With the rise in the number of papers on azole antifungal design and discovery, a more in- depth understanding the most recent and authentic information about this class of drugs might be beneficial. To this end, we for the first time summarized the state-of-the-art information about azole drugs, with a specific focus on those in the pipelines of pharmaceutical companies, into four generations with regard to their structural similarity. More importantly, this review highlights information on the structure activity relationship (SAR), target description, hybrid antifungal agents as possible future generation, and other useful issues to streamline research towards designing new efficient azole antifungal structures in future. 1. Introduction 1.1. Fungal infection The Fungi kingdom is one of the most diverse organismal kingdoms on the planet. Unlike the bacteria, fungi have eukaryotic cellular functions, making them more closely related to human than bacteria [1]. Out of more than two million fungi species, about 600 types are known as human fungal pathogens, and only 3–4% of these species account for > 99% of invasive fungal infection (IFI) [2]. These types of fungal infections are often life-threatening and have a higher mortality rate in comparison to superficial fungal infections, which are common in human beings [3–5]. A particularly susceptible population for the development of IFIs are individuals with compromised immune function such as those with organ transplants and AIDS (acquired immune deficiency syndrome) as well as those who are on prolonged immunosuppressive medications such as corticosteroids. The patients with antibiotics, cancer che­ motherapies, and the ones in intensive care units (ICU) are also at greater risk to develop IFI [4]. This sheds some light on the opportu­ nistic nature of fungal infection [6]. Over the last 30 years, IFIs in­ creased with the rising number of the vulnerable population and an­ nually caused 1–2 million deaths. The mortality rate was much greater than that of malaria or tuberculosis [7–9]. > 90% of the deaths are attributed to Candida and aspergillus species [10]. Candida species, in­ cluding Candida albicans, Candida parapsilosis, Candida krusei, Candida glabrata, and Candida tropicalis have emerged as invasive candidiasis, whose mortality rate is increased up to 75% as they usually affect re­ spiratory, gastrointestinal, skin, and urogenital tracts [11–13]. More­ over, aspergillosis along with aspergillus spp. such as fumigatus, flavus, niger, terreus, and parasiticus mainly affect external ear, lungs, eye, and brain, with a mortality rate of 50–90% [14]. Although Fungi infection is the first scientifically reported or­ ganism-derived disease, prior to bacterial infections [15], antifungal drug discovery has mightily lagged behind antibacterial chemotherapy, resulting in one of the main problems with targeting fungal species, i.e. the eukaryotic nature of fungi makes them intrinsically difficult to be targeted without the probable toxic effects of antifungal drugs in https://doi.org/10.1016/j.bioorg.2020.104240 Received 9 June 2020; Received in revised form 17 July 2020; Accepted 11 August 2020 ⁎ Corresponding authors at: University of Arizona college of medicine Phoenix and Pima college, Tucson, Arizona, USA. (Mehrnoosh Hashemzadeh). Department of Medicinal Chemistry, Faculty of Pharmacy & The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran (Alireza Foroumadi). E-mail addresses: mhashemz1@yahoo.com (M. Hashemzadeh), aforoumadi@yahoo.com (A. Foroumadi). Bioorganic Chemistry 104 (2020) 104240 Available online 28 August 2020 0045-2068/ © 2020 Elsevier Inc. All rights reserved. T
  • 2. human cells [15]. Accordingly, the development of selective antifungal drugs with lower side-effect is a hotbed of research among medicinal chemists [16]. 1.2. Current medications Currently, there are several well-known mechanisms to treat sys­ temic and superficial fungal infections using roughly 40 branded anti­ fungal drugs in the market and the other drug candidates in clinical trials, which adopt new or convenient mechanisms to target fungi (Fig. 1) [2,17–20]. Interestingly, the previous studies have introduced some new tar­ gets to combat fungal infections, hoping that the emerging strategies may yield the treatment for resistant fungal infections (Fig. 2) [21–24]. Despite new research and development in this field, there is still no new approved drug for the more recent strategies. As shown in Figs. 1- 14-α demethylase inhibitors (azoles) are the most important and the most approved clinical candidate drugs. According to previous studies, classified azoles cover several char­ acteristics of the most potent antifungals. Over the past half-century, this family has had a significant effect on controlling invasive and su­ perficial fungal infections. Their newer generation further increased highly desired properties such as safety, lower risk of drug-drug inter­ actions, proper pharmacokinetics, and improved activity spectrum [25–27]. With the large number of azole antifungals in the clinics and in experimental medicinal chemistry institutions, keeping track of pro­ mising molecules that are under development is challenging for the researchers in the field. In this paper, we provide a comprehensive review and an update of all approved and clinical candidates of azole antifungal drugs, as azole pedigree in regard to their structural simi­ larities. Herein, the description of the target, salient medicinal chem­ istry points regarding azole structures (SAR), and the selectivity of these structures for the concerned target are discussed. Furthermore, hybrid antifungal agents as a prospective future generation are introduced to pave the way for developing new efficient azole antifungal structures in the future. 2. Azole family 2.1. History and pedigree The first report on the antifungal activity of an azole compound was presented by Woolley (1944), a serendipitous discovery that declared the antimycotic effect of benzimidazole moiety for the first time [28]. Afterwards, several other researchers indicated other azole antifungal structures such phenethylimidazole [29] and substituted benzimida­ zoles [30]; however, none of these reports led to the development a drug until 1958, when chlormidazole (1, Fig. 3) was introduced, the first azole drug marketed by Chemie Grünenthal. Accordingly, chlor­ midazole was the pioneer of enormous research on azole antifungal activities. Currently, there are about 40 azole-containing drugs and drug candidates, which could be classified into more than three gen­ erations [15,31,32]. This field of study is still of great interest. 2.1.1. First generation The first generation of azole family emerged with the introduction of three compounds: clotrimazole (13) from Bayer AG in 1969, Janssen’s miconazole (2) at the same time, and later econazole (3) in 1974. These three drugs were launched as topical antifungal agents making azoles an indispensable asset in treating fungal infections[15]. Following the discovery of these early structures, several analog com­ pounds are marketed, which can be categorized into the first-generation group due to their similar physiochemical properties and indications in treating superficial fungal infections such as dermatomycoses, tinea versicolor, cutaneous, and vaginal candidiasis as well as topical for­ mulations. Considering their structures, they have similar imidazole- contained backbone and could be further sub-divided into three cate­ gories: miconazole-based structures, clotrimazole- based structures, and vinyl-imidazole derivations, as shown in Fig. 4. Fig. 1. Antifungal drugs. the number of approved and clinical trial agents are summarized. M. Shafiei, et al. Bioorganic Chemistry 104 (2020) 104240 2
  • 3. Miconazole (2) has well-documented activity in the treatment of oropharyngeal candidiasis as well as vulvovaginal candidiasis, tinea corporis, tinea pedis, and tinea cruris [33]. It also has a fungicidal ac­ tivity via direct membrane damage. Clinically, miconazole adminis­ tration routes are topically or intravenously; however, miconazole in­ fusion is also associated with adverse reactions such as nausea, fever, and more importantly cardiotoxicity when rapidly infusing that was due to a formulation containing Cremophor® EL because of its low so­ lubility [34]. All the miconazole analogs are derived from a key inter­ mediate, 2-(imidazolyl)-acetophenone. The only exception is butoco­ nazole (11), which is derived from 4-(4-chlorophenyl)-1-(1H-imidazole- 1-yl) butan-2-ol structure. According to FDA, clotrimazole (13) is safe and effective for the prevention and treatment of oropharyngeal, vulvovaginal, and cutaneous candidiasis and the other superficial dermatologic infections [35]. Clotrimazole (13) synthesis was reported three years after its de­ velopment. Benzophenone derivations could be used for clotrimazole (13) and bifonazole (14), and dibenzosuberone for eberconazole (15) synthesis. Becliconazole (16) is a new clotrimazole-like compound, which is recently entered into the clinical trials from Menarini Company. Luliconazole (21), the newest first-generation azole approved by FDA, contains a unique vinyl-imidazole structure. It has a broad-spec­ trum topical effect against dermatophytes such as Trichophyton rubrum, Microsporum gypseum and Epidermophyt on floccosum [36]. In this group, TS-80 (22) entered into phase-I clinical trial from Shikoku company and revealed an activity against cutaneous fungal infections; however, this trial has not yet been completed. 2.1.2. Second generation For second generation azole antifungals, there are three main changes in the structure of the compounds which resulted in many improvements in terms of the safety, spectrum of action, and pharma­ cokinetics of azole drugs. It should be noted that following the second generation of azoles, subsequent generations are just modifications and analogs of these structures. In this regard, the main changes are evident in Janssen's (1981) antifungal ketoconazole (23), the first drug containing a heterocyclic dioxolane functional group, replacing the ketone with a new long-tail Fig. 2. New fungal cell targets. (1) Fig. 3. Structure of chlormidazole (1). M. Shafiei, et al. Bioorganic Chemistry 104 (2020) 104240 3
  • 4. structure; however, the imidazole moiety remained similar to its pre­ decessor. This was the first orally active, broad-spectrum azole to treat systemic mycoses [37]. Although ketoconazole (23) has several drawbacks resulting in a black box warning from the US FDA, it was preferred to amphotericin B (AmB) for non-life threatening systemic fungal infections until 1990 [38]. One of the major problems with ketoconazole (23) is hepato­ toxicity caused by CYP450 inhibitions, indicating the imidazole-con­ tained lower selectivity for fungal compared to mammalian P450 en­ zymes. The other prevailing problems are bioavailability and pharmacokinetic interactions, which finally led to removal of this drug from the FDA list of approved medication to treat systemic mycosis in 2013 [39]. The third and the most important breakthrough in the development of azole structure was the introduction of a triazole ring replacing the bygone imidazole moiety in the azole structures. Triazole was first used along with fluconazole (24) developed by Pfizer (1989) [40]. The su­ perior advantages of fluconazole (24) were selectivity for fungal CYP enzyme over mammalian, a larger spectrum of action, increased metabolic stability, lower protein binding, enhanced bioavailability, greater water solubility to be used as intravenous indications, high CNS penetration, and lower side-effects [41–43]. Interestingly to achieve higher IV bolus administration of fluconazole (24) in clinics, even greater solubility was necessary. This problem was solved using a phosphate ester of fluconazole (24) which was approved in 2003 [44]. Fosfluconazole (28) could be active with the action of alkaline phos­ phatase enzymes [45]. The discovery of fluconazole (24) is one of the most interesting medicinal chemistry studies on the modification of a structure to achieve desirable properties (Fig. 5) [46]. In its synthesis, an epoxide ring (Fig. 6A) is the key intermediate achieved by well-known Corey- chaykovsky epoxidation with trimethyl sulfoxonium iodide (TMSI) re­ agent employed as the ring closer [47–50]. At the same period, another important triazole-based structures improved by Janssen pharmaceuticals were terconazole (25) and itra­ conazole (26) [15], both of which had a long-tail structure to similar that of ketoconazole (23) as well as similar low bioavailability. They have several side-effects, including cardiotoxicity. Although they have a Fig. 4. First generation azole structures: A: miconazole (2) analogs, B: clotrimazole (13) analogs, C: Vinyl-imidazole structures. M. Shafiei, et al. Bioorganic Chemistry 104 (2020) 104240 4
  • 5. broader spectrum of antifungal activity in comparison to ketoconazole (23) and fluconazole (24), they couldn’t be an alternative to fluconazole (24) in many cases [51–53]. Itraconazole (26), however, was preferred to ketoconazole (23) in the treatment of mycoses, Blastomycosis and invasive aspergillosis [54,55]. In terms of synthesis, itraconazole (26) and similar structures utilize a common route through dioxolane in­ termediate formation at four major steps (Fig. 6B) to form the targeted compound [37].(See Table 1) When the first-generation structures were compared to the second- generation (Fig. 7), the latter generation improved upon the spectrum of activity and added routes of administration; however, there were still challenges regarding pharmacokinetic and safety profiles. Conse­ quently, the third generation of azole drugs were the improved struc­ tures of the second generation and could be sub-divided into two groups of fluconazole (24) and itraconazole (26)-like structures. Table 2 shows the major physiochemical and pharmacokinetic properties of the main second-generation azole drugs [45,56,57]. 2.1.3. Third generation With the increasing concerns about the high incidence of fungal infections, new structures emerged as the third-generation azoles or triazole generation. These drugs are improved structures of the second- generation antifungals divided into two groups of fluconazole (24) and itraconazole (26) like structures (Fig. 8). In both of these groups of the third generation, major medicinal chemical efforts were made to im­ prove pharmacokinetic properties and safety profile, extend the spec­ trum of activity, and, more importantly, design a structure to combat resistant types of fungal infections. In Table 3, major physiochemical Fig. 4. (continued) M. Shafiei, et al. Bioorganic Chemistry 104 (2020) 104240 5
  • 6. and pharmacokinetic properties of several third generation drugs are listed [58–67]. Since fluconazole (24) is inactive against molds such as Aspergillus spp and increases the resistance of Candida spp, in 2002, the FDA ap­ proved voriconazole (30) produced by Pfizer with a fluoropyrimidine ring instead of the second triazole of fluconazole (24) [68]. This structural difference has made the compound > 30 fold more potent for C. albicans, 20 fold more potent for C. glabrata, and completely active against all Aspergillus spp [69]. In 2004, the Schering Plough Research Institute working on a new lead, SCH51048, observed that antifungal activity was higher than the hypothesized concentration, suggesting the presence of an active metabolite. Following LC-MS/MS analysis, a hydroxylated metabolite was discovered. By using this as a lead and after synthesis of all side- chain monohydroxylated diastereomers, posaconazole (31, SCH56592) was discovered as a new triazole antifungal agent, providing a prime example of active metabolite-based drug design (Fig. 9) [70,71]. Posaconazole (31) showed a broader spectrum of activity against all fungus organisms covered by other new broad-spectrum antifungal drugs. Moreover, it had an additional activity against Zygomycetes, Scedosporium spp, and also was effective in the treatment of CNS fungal infections and cryptococcal meningitis [72]. The FDA approved posaconazole (31) for prophylaxis of invasive Aspergillus or Candida infections in hematopoietic stem cell Fig. 4. (continued) Fig. 5. Fluconazole (24) structure optimizations. M. Shafiei, et al. Bioorganic Chemistry 104 (2020) 104240 6
  • 7. transplantation (HSCT) recipients with graft versus host disease (GVHD), as well as in patients with hematologic malignancy with prolonged neutropenia and oropharyngeal candidiasis [73–75]. Efinaconazole (32), as a new topical triazole antifungal for the treatment of mild to moderate onychomycosis, received its first global approval on October 3, 2013 in Canada by Valeant Pharmaceuticals International. This compound has a better safety profile compared with the oral treatment of onychomycosis [76]. In cases of intravenous formulations of posaconazole (31), vor­ iconazole (30), and itraconazole (26), cyclodextrin is added to increase solubility, which is of concern due to potential nephrotoxicity [77]. This concern was remedied by the medicinal chemistry work of Basilea Pharmaceuticals and Astellas Pharma. Isavuconazonuim sulfate, a water-soluble prodrug of isavuconazole (33) containing a 1-[N-methyl- N-[3-[2-(methylamino)acetoxymethyl]pyridyn-2-yl]amino carbony­ loxy] ethyl substituent, easily converts to its active form by the action of plasma esterase (Fig. 10). Therefore, the oral and intravenous for­ mulation no longer needs cyclodextrin for enhanced solubility [78,79]. (See Fig. 11) As a broad-spectrum azole drug, isavuconazole (33) has an anti­ fungal activity profile similar to that of voriconazole (30). In 2014, the FDA approved it for treatment of invasive aspergillosis [80]. Another example of triazole antifungal in clinical trials is ravuco­ nazole (34), with a slight difference in structure compared to isavuconazole (33), but with a similar antifungal profile to that of voriconazole (30). Ravuconazole (34) is active against fluconazole (24) and itraconazole (26) resistance infections. It was discontinued from phase II for aspergilusis and candidiasis in August 2007, and was re­ placed with fosravuconazole bis(L-lysine) created by Bristol-Myers Squibb. The first global approval of this prodrug of ravuconazole (34) with the brand name of NAILIN® as 100 mg capsules was for onycho­ mycosis in Japan on July 27, 2018 [81]. Albaconazole (35) with a 7-chloro-quinazoline-4-one structure bears more potent antifungal in vitro activity than Amphotericin B. Furthermore, a wide spectrum of action and favorable pharmacokinetic with safety profile [82] was originated by Grupo Uriach. In 2013, Al­ lergan (Actavis Inc) picked up the worldwide rights for albaconazole (35) from Palau Pharma S.A. Other new compounds of this generation are also currently in phase- III, such as genaconazole (36) and iodiconazole (37), phase II such as saperconazole (39), ICI-D0870 (42) and UR-9751 (41), and phase I such as embeconazole (38), DUP-860 (40), SS-750 (44), SSY-726 (46) and PC-945 (47). Moreover, some new structures like UR-9660 (43) and SDZ-89–485 (45) are in preclinical stages [18–20,83]. Many triazole alcohol derivations have been investigated and sev­ eral new and known scaffolds incorporated into their structures are assessed in terms of their antifungal activity [84–86]. This new tech­ nique for antifungal agent discovery is highly growing in medicinal Fig. 6. Second generation azole drugs intermediates synthesis, A: The intermediate of Iitraconazole (dioxalene) synthesis roots. B: Fluconazole’s intermediate (epoxide) synthesis root. M. Shafiei, et al. Bioorganic Chemistry 104 (2020) 104240 7
  • 8. chemistry research [87–95]. 2.1.4. Fourth generation In the previous generations, new structures successfully emerged and solved the resistance, potency, the spectrum of action, and phar­ macokinetics problems of azole family. One of the main problems, known as the ‘Achilles heel’ for azoles, is the drug-drug interactions caused by probable human CYP3A4 enzyme inhibition [33]. This pro­ blem is a concern since many patients suffer from complicated conditions such as AIDS, cancer, and impaired immune function and need polypharmacy. The metal binding region in azole structures is the main part of the inhibitory effect as for human CYP3A4 inhibition. Following the replacement of imidazole with triazole ring, the se­ lectivity increases, even though, the problem still exists. Successful ef­ forts have been made in Viamet Pharmaceuticals, Inc. (Durham, NC, USA). Using an appropriate medicinal chemistry strategy, they in­ troduced three novel azole structures (Fig. 10) with high selectivity for fungal CYP51 and increased potency and desirable pharmacokinetics and physiochemical properties (Table. 4), in comparison to the other structures [96–98]. Replacement of high-avid metal binding and more potent triazole ring with less-avid metal binding and less potent tetra­ zole ring and the compensation of low potency with modification of side-chain were performed to have the more selective and potent fourth-generation of azole structures [99,100]. They used voriconazole (30) for its lower anti-target (CYP3A4) af­ finity compare to the other azoles, as a leading compound to design new compounds. Quilseconazole (48, VT-1129) is now in phase I clinical trial in cryptococcal meningitis. It has had a promising efficacy in the murine model and increased one-month survival, compared to fluco­ nazole (24). In Jun 2016, VT-1129 (48) received Fast Track orphan drug designation for the treatment of cryptococcal meningitis [PO] (in volunteers) in the USA [101,102]. Table 1 First generation azole information’s about important physiochemical proper­ ties. Generic name Physiochemical properties CLogP1 tPSA2 LogS3 Becliconazole 4.83 24.83 −5.94 Bifonazole 4.74 15.6 −6.28 Butoconazole 6.86 15.6 −7.56 Clotrimazole 5 15.6 −6.97 Croconazole 4.5 24.83 −5.68 Dapaconazole 5.26 24.83 −6.64 Eberconazole 4.97 15.6 −6.1 Econazole 5.1 24.83 −6.39 Fenticonazole 6.72 24.83 −8.23 Flutrimazole 4.57 15.6 −6.75 Isoconazole 5.8 24.83 −7.08 Luliconazole 3.49 39.39 −5.54 Miconazole 5.8 24.83 −7.09 Neticonazole 4.24 24.83 −5.48 Omoconazole 6.06 34.06 −7.44 Oxiconazole 6.46 37.19 −7.69 Sertaconazole 6.16 24.83 −7.45 Sulconazole 6.27 15.6 −7.02 Tioconazole 4.78 24.83 −5.76 Ts-80 5.52 32.6 −6.98 Zinoconazole 5.08 39.9 −6.198 1. Calculated octanol–water partition coefficient, 2. Topological polar surface area, 3. Aqueous solubility Fig. 7. Second generation azole drugs structures. Table 2 Major physiochemical and pharmacokinetic properties of important second generation azole drugs. Generic name Physiochemical properties Pharmacokinetic properties CLogP1 tPSA2 LogS3 PPB (%)a Half-life (h) Vd (L/ Kg)b Fluconazole −0.44 76.15 −2.127 12 31 0.7 Itraconazole 5.98 98.04 −9.752 > 95 64 11 Ketoconazole 3.63 66.84 −6.421 > 95 8 No data 1. Calculated octanol–water partition coefficient, 2. Topological polar surface area, 3. Aqueous solubility a: plasma protein binding, b: volume of distribution M. Shafiei, et al. Bioorganic Chemistry 104 (2020) 104240 8
  • 9. Oteseconazole (49, VT-1161) another tetrazole compound after successful phase III trial for vulvovaginal candidiasis in March 2019 (PO, Capsule) (NCT03840616), the Mycovia Pharmaceuticals and Jiangsu Hengrui Medicine have teamed up to develop and market the first FDA approved drug for vulvovaginal candidiasis [103,104]. VT- 1598 (50), with the broadest spectrum of antifungal activity among tetrazoles, is the most recent compound from the tetrazole group [105], whose important anti-coccidioidomycosis effect (in vitro MIC: 0.06–0.5 mg/L) import this compound to preclinical development in USA (PO) [106]. Another multidrug resistance fungal spices, Candida auris, is also affected by VT-1598 (50) with MIC of 0.03–8 mg/L in vitro [107]. Following the efforts of Viamet pharmaceuticals, the significance of side-chain optimization in increasing the potency and metal-binding Fig. 8. Third generation azole drug structures (blue structures derived from fluconazole and red structures derive from itraconazole). (For interpretation of the references to colour in this figure legend, the reader is referred to the web version of this article.) M. Shafiei, et al. Bioorganic Chemistry 104 (2020) 104240 9
  • 10. group tetrazole in having better selectivity became obvious. This new family and all other new structures in clinical trials showed that an old mechanism could be a new hope. 2.1.5. Hybrid structures as future azole-generation The hit to lead process and further development to achieve a drug candidate is time and cost consuming [108]. Classical combinatorial chemistry or modern virtual screening approaches have not yet brought a breakthrough in the discovery of novel drug candidates [109]. In order to obtain new antifungal drugs that are affordable and able to avoid the emergence of resistant strains, hybrid drug discovery comes to mind. Molecular hybrids of biologically active structures are now gaining momentum worldwide and may be a key tool in the arsenal of medicinal chemistry efforts for drug discovery [110]. Hybrid or dual- acting molecules are defined as combination of two or more chemical scaffolds that act at different targets to create more specific and Fig. 8. (continued) M. Shafiei, et al. Bioorganic Chemistry 104 (2020) 104240 10
  • 11. powerful drugs [111]. Fluconazole (24) as a favorable antifungal drug due to its worthy in vivo efficacy and pharmacokinetic property, has been attracted for hybrid drug discovery. Fluconazole analogues obtained by facile replacing one triazole ring by other potential antifungal scaffolds di­ rectly or by using suitable linkers [112]. This new way to design structures makes fluconazole hybrids potential future generation of azole antifungals. The triazole nucleus especially 1,2,3-triazole ring has been utilized as a linker in various antifungal drug hybridizations by means of click reaction [113]. Several research groups introduced 1,2,3-triazole ring in the side chain of azole structure to design hybrid structures and survey antifungal effects [90,114–118](Fig. 15). This motif act not only as a linker, but also interacts efficiently with active site of CYP51 providing in many cases improved physiochemical properties of struc­ tures [119]. Recently medicinal chemists have made considerable efforts to de­ sign potent hybrid antifungal structures of fluconazole, some of which are summarized (Fig. 12). Natural products are a dominant source of biologically active structures and hybridization of these structures with fluconazole can lead to potent drugs [108]. Zhang et al, [89] report design and synthesis of novel series of carbazole-triazole conjugates. Carbazole derivations as naturally occurring phytochemicals are known for pi-conjugated Table 3 Major physiochemical and pharmacokinetic properties of several 3rd genera­ tion azole drugs. Generic name Physiochemical properties Pharmacokinetic properties CLogP1 tPSA2 LogS3 PPB (%)a Half-life (h) Vd (L/ Kg)b Voriconazole 0.52 72.91 −2.72 58 6 4.6 Posaconazole 4.1 109.04 −8.82 > 95 25–31 428 Efinaconazole 2.14 51.43 −2.97 – 29.9 – Isavuconazole 2.68 84.34 −5.19 98 56–77 6.5 Ravuconazole 2.68 84.34 −5.2 98 76–202 10.8 Albaconazole 2.14 80.86 −4.72 98 30–56 Very large 1. Calculated octanol–water partition coefficient, 2. Topological polar surface area, 3. Aqueous solubility a: plasma protein binding, b: volume of distribution Fig. 9. Active metabolite of early lead SCH51048, posaconazole (31, SCH 56592). M. Shafiei, et al. Bioorganic Chemistry 104 (2020) 104240 11
  • 12. motifs that could inhibit topoisomerase, intercalate with DNA, and also disrupt the integrity of the microorganism membrane [120]. Their ef­ forts led to compound A (Fig. 12) with MIC = 2–32 µg/mL against most of the tested fungal strains. Compound A also, intercalates into DNA to possibly block DNA reproduction. Elias et al [121] in an effort for synthesis new azole hybrids with coumarin and quinolone scaffold showed that their coumarin hybrids antifungals (compound B, Fig. 12) could penetrate into endoplasmic reticulum of fungal cells where, true target CYP51 generally exist in. This potentially can enhance the effi­ cacy of azole antifungal drugs. Combination of known antifungal, antimicrobial, and anticancer drugs to azole structures are also among high interest. This hybridiza­ tion will help to combat resistance strains due to the possibly different mechanism of actions. Experiments conducted by Sheng et al, [122] elucidate design and synthesis of the first CYP51/HDAC dual inhibitors. They successfully developed potent antifungals agents with activity against azole-resistant clinical isolates. The HDAC family of proteins are key enzymes involved in gene regulation, cell proliferation, and are known anticancer targets. Vorinostat (SAHA) (Fig. 12) an HDAC in­ hibitor, was previously confirmed to possess synergistic effects with fluconazole (24) against azole-resistant strains. Inspired by these find­ ings, new anti-fungal dual inhibitor hybrids emerged. Compound C, with MIC = 0.25–0.5 µg/mL and potent in vivo efficacy against azole resistance candidiasis was introduced as the best CYP51/HDAC hybrid. Before this, they also designed a hybrid of rosiglitazone and other de­ rivations of thiazolidinedione with fluconazole (Fig. 12) and investigated anti-fungal activities. Their target compound D, with MIC values against C. albicans in the range of 0.03–0.15 µM, proved the efficiency of dual inhibitors in antifungal drug discovery [111]. Another team, studied the antifungal activity of 5-flucytosine combination with fluconazole (123, Fig. 12). Two potent antifungals combined in one molecule. Their efforts led to compound E with MIC = 0.008 and 0.02 mM against C. albicans ATCC 90,023 and clinical resistant strain C. albicans respectively. They also showed that new hybrids could permeate membrane of C. albicans and also could inter­ calate into calf thymus DNA helix and block DNA replication by making a steady supramolecular complex [123]. 2.2. Mechanism of inhibition The recognition of a bottleneck for efficient antifungal drug dis­ coveries based on the eukaryotic nature of fungi and its similarities with the human is necessitous. Similar to most of the other drugs, the be­ ginning of azole class was by chance, and none of the antifungal drugs were discovered using rational target-based drug design strategy. One of the main targets of small molecules effective in combating fungal infection is cell membrane and the biosynthesis of ergosterol, a derivative of mammalian cholesterol. Ergosterol has several functions in fungi, the important of which is its role in cellular proliferation. To this end, a planar a-face (no methyl group at C14) structure and a double bond in C5-6 are required to have these structural character­ istics. There are several important enzymes in the biosynthesis pathway Fig. 10. Activation of Isavuconazonium sulfate prodrug to its active drug. M. Shafiei, et al. Bioorganic Chemistry 104 (2020) 104240 12
  • 13. [124], one of the predominant enzyme in required is CYP51 (Lanestrol- 14α-demethylase) (ERG-11) from cytochrome P450 family, which cat­ alyzes an oxidative removal of lanosterol, 14-methyl group and leads to a 14–15 double bond (Fig. 13) [125]. In the active site of CYP51, an iron-containing porphyrin provides oxygen for this first oxygen-de­ pendent reaction in the ergosterol biosynthesis. Recently, several pa­ pers fully-described the mechanistic details of CYP51 action [126–129]. Furthermore, a recent study showed that CYP51 might be involved in distinct process of eluding human immune system, a finding that will likely grow with the increase with the growth of research. [130]. Thus, accumulation of methylated sterols would cause a fungistatic effect in yeast and fungicidal effect in molds in most of the azoles, except in high doses, and voriconazole (30) and itraconazole (26) (they are considered to have fungicidal inhibition). Pertaining to immunocompromised pa­ tients, the eradication of the fungal infection is of essence and fungi­ cidal effects in therapeutic doses are highly beneficial [53]. Thus, knowing the enzyme structure is crucial for efficient inhibition of CYP51 and designing selective and potent inhibitors. Elucidation of the CYP51 structural information was challenging before obtaining a complex of CYP51 orthologue from prokaryote myco­ bacterium tuberculosis (carrying about a 33–35% sequence homology to that fungal) with fluconazole (24). The reason is that purification and crystallization of a highly hydrophobic membrane-bound protein is arduous. Information from other CYP51 orthologues is not useful. For ex­ ample, VFV, as a potent anti T. cruzei drug candidate, today, has a weak inhibitory effect on fungal CYP51. Moreover, VNI, as another anti- Chagas disease drug candidate with an important H-bond interaction of its carboxamide group with the T. cruzei active site, does not form any H-bond in fungal active site protein [131–133]. However, biochemistry works and X-ray crystallographic studies have recently developed a protein expressed in some organisms like Escherichia coli, S. cerevisiae, C. albicans, and C. glabrata and also in other important species [129,134]. Fig. 11. Fourth generation azole drugs structures derive from voriconazole. Table 4 Major physiochemical and pharmacokinetic properties of 4th generation azole drugs. Generic name Physiochemical properties Pharmacokinetic properties CLogP1 tPSA2 LogS3 PPB (%)a Half-life (h) Vd (L/Kg)b Quilseconazole 4.99 82.14 −6.59 No data > 6 days No data Oteseconazole 4.67 82.14 −6.64 No data > 48 1.4 VT-1598 5.73 105.93 −7.91 > 95 22 No data 1. Calculated octanol–water partition coefficient, 2. Topological polar surface area, 3. Aqueous solubility a: plasma protein binding, b: volume of distribution M. Shafiei, et al. Bioorganic Chemistry 104 (2020) 104240 13
  • 14. From many studies of azole binding site interactions in the ligand- binding pocket of CYP51 in different species reported by diverse groups, several common interactions have emerged with conserved amino acids in the active site. The most conserved domain is heme- binding pocket, and also motifs that donate to set the heme into the core structure and some other residues on the surface of the active site [135]. Recent biochemistry studies declare that the ligand-binding pocket (LBP) in different species such as S. cerevisiae, C. glabrata, and C. albicans has similarities with the structural and chemical characteristics of lanosterol demethylase protein [135]. Azole drugs inhibit the en­ zyme reaction by non-competitive reversible interaction with the heme and prevent accessing of protons to the active site. There are several similar interactions with other residues. For example, structures have shown that C. albicans CYP51 protein complex with posaconazole (31) or oteseconazole (49) has similar binding interactions with 22 amino acids, on which oteseconazole (49) has an important H-bond with the His-377 from its trifluoroethoxyphenyl oxygen (Fig. 14). His377 is highly conserved in different fungal pathogens and also is specific for fungi and not humans. This shows the importance of H-bonding in the fungal CYP51 active site complex with inhibitors for potent and selec­ tive drug design. Key binding pocket interactions for fluconazole (24) in LBP of S. cerevisiae and oteseconazole (49) in C. albicans CYP51 active site are presented in Fig. 14 [130,134]. Despite these similarities in the CYP51 amino acid sequences and Fig. 12. Some of important fluconazole hybrid antifungal agents. M. Shafiei, et al. Bioorganic Chemistry 104 (2020) 104240 14
  • 15. interactions of different inhibitors with them, allowing for development of selective inhibitors with a wide spectrum of action in CYP51 human pathogens, it is necessary to study the conformation and binding in­ teractions of each new inhibitor. This will permit and allow investiga­ tion of target-driven information in the context of target-based drug discovery, setting new interactions on selective and non-conserved re­ sidues to reduce the risk of resistance in future [136,137]. Significant progress has been made with regard to information on these X-ray crystal structures in characterization of CYP51 for anti­ fungal drug discovery. Several complex structures of CYP51 with in­ hibitors are available from RCSB Protein Data Bank (PDB) (rcsb.org). The information on CYP51 structures is presented in Table 5 [127,134,136,138–142]. 2.2.1. Structure activity relationship (SAR) After the voriconazole (30) discovery, there were > 1000 flucona­ zole-derived structures that were synthesized and experimentally tested at Pfizer, an inefficient and costly process [143]. Therefore, to design and survey new azoles, as a leading class of important antifungal drugs, a good model of structure–activity relationships could be cost and time beneficial. From many studies on azole structure–activity relationships and others surveying interactions in target protein with x-ray crystal­ lography or efficient docking methods, several useful pieces of in­ formation have been obtained. Here, we gathered, modeled and sum­ marized important structural features for future studies in the field. In general, all azole drugs have a similar structural model (Fig. 15), consisting of a Heme binding group, a three-atom linker, side-chain A and side-chain B that are fully described below. - Heme binding group: This ring is crucial for the antifungal activity of the azole group without any substitutions that bind to the linker with N-1 atom. The basic N-3 of the imidazole ring or N-4 of the triazole or tetrazole ring is positioned vertically to the porphyrin plane, as the iron of the active site with the length of the coordinated bond of 1.2–2.1 A⁰. The basicity of heteroaromatic nitrogen-containing rings does not have any re­ levance with the inhibitory potency of the azole structure; however, it is directly associated with liver toxicity and drug-drug interactions. We can also define basicity as iron-binding affinity. Thus, tetrazole with the lowest basicity (-1.1 vs 2.3 for triazole and 6.8 for imidazole) has minimum inhibitory for CYP450 and related adverse effects [99,128,144]. - The three-atoms linker: Although we named this linker as a three-atom linker, it could be varied between 1 and 4 atoms. This three-atom shape makes a special distance between different arms of structure, leading to greater potency [145]. The atom number 1 of the linker group in azole structures is commonly carbon and does not have any substitutions except topical agents such as clotrimazole analogs and vinyl imidazole derivations. For the atom number 2 and 3 in the linker, which are mostly a carbon, chirality is important for antifungal activity [146,147]. Presence of an oxygen atom as a hydroxyl group on C-2 has several beneficial effects such as enhanced potency due to an indirect H-bond via water mole­ cules with active site, improved pharmacokinetics properties and water Fig. 13. A. Sterol biosynthesis from Acetyl-CoA to ergosterol in fungi. B. The active site of CYP51[PDB: 4LXJ]: lanosrterol (green), heme (cyan), iron (orange), oxygen(red) and Reaction steps catalyzed by C14-demethylase. (For interpretation of the references to colour in this figure legend, the reader is referred to the web version of this article.) M. Shafiei, et al. Bioorganic Chemistry 104 (2020) 104240 15
  • 16. solubility, better tolerated and stable metabolism. Moreover, it is needed to design prodrugs such as phosphate ester of fluconazole (24), voriconazole (30) and ravuconazole (34) [44,148,149]. A methyl group on C-3 could enhance potency against moulds. The chiral form gives special conformation that side-chain B moves in antiperiplanar to side- chain A and also this methyl group fills the binding pocket of C-13 of lanosterol [46,150]. Other substitutions such as CH (double bond with C-3) and two methyl or two F (non-chiral C-3) have also the desired activity. In some of the first generation structures, C-3 became an oxygen, sulfur or NR2 (oxime and hydrazine) (miconazole (2) and its derivations), which maintains antifungal activity and also conforma­ tional restriction of structure with a ring on C-2 and C-3, such as dioxalene (itraconazole (26) or ketoconazole (23)) or tetrahydrofuran (posaconazole (31)); however, they do not enhance potency, or the other pharmacokinetic properties and other groups such as ester, amide, and carbamate are not appropriate for this replacement in an­ tifungal activity, in contrast to antitrypanosome cruzi and anti­ tubercular activity [151,152]. - Side chain A: Side chain A in the structure that is always a halogenated phenyl, is essential for inhibitory effect and appears to be located in the same hydrophobic tunnel occupied with the 17-alkyl chain of lanosterol. It Fig. 14. A: 3D structure of S. cerevisiae CYP51 complexed with fluconazole in the active site (PBD: 4WMZ). B: 3D structure of C. albicans CYP51 complexed with oteseconazole in the active site (PBD: 5TZ1). Cyan dash line: metal coordination, Red dashes lines: water-mediated hydrogen bonding, green dash line: pi-stacking interaction. M. Shafiei, et al. Bioorganic Chemistry 104 (2020) 104240 16
  • 17. has pi-pi stacking interactions with several residues like Y132 (a con­ served amino acid among fungus). Substitutions of the phenyl ring in all carbons are tolerated except in groups larger than chlorine in C-2 and C- 6 that have unfavorable steric clashes and cause a decrease in the binding affinity of inhibitors in the active site. A greater potency is obtained from fluorine atoms in C-2 and C-4. However, other large bulky groups such as 4-fluorophenyl could be replaced in para position and maintain the CYP51 inhibition activity of molecules [145,153]. - Side chain B: Side chain B area is similar to that of the heme-binding group as we could design potent and favorable pharmacokinetic properties only with optimization in this side chain. Many medicinal chemistry studies survey various substitutions in this side chain as can be seen in third- generation and hybrid structures (Fig. 7 and Fig. 12) [112]. In general, a linker binds the side chain B to C-3 of tree atom linker, and antifungal activity differs from several different types of linkers here. Studies evaluating this area show that ether, thioether, carbon (–CH2-, -CHR-, -CR2-), -NR2, -SO2-, heteroaromatic or hetroaliphatic (e.g., piperazine, piperidine) rings are well-tolerated and are good types of linkage, while amide, ester, and -NHR reduce antifungal activity [154–156]. Substitutions binding to the structure via the mentioned linkers could make several hydrophobic, steric and important H-bond interactions in the deep binding cleft of target protein. Before any de­ scription of this area, we divided azole drug into two groups based on the size of the side chain, short-tail and long-tail structures. For short tail structures, the linker may or may not have small substitutions, like fluconazole (24), ravuconazole (34), and vor­ iconazole (30) structures. Small hydrophobic, electron-rich and elec­ tron-withdrawing groups (e.g., halogens, –CN, and halogenated het­ eroaromatic rings) enhance potency in the ortho and para-position of a phenyl or heteroaromatic ring attached to the linker, with probable steric and hydrophobic interactions (both aromatic ring and its sub­ stitution). In the voriconazole (30) case, fluorine on the pyrimidine ring and Tyr122 of A. fumigatus form an H-bond interaction. Meta-position substitutions are not favorable for activity [128,155]. For long-tail structures like itraconazole (26), posaconazole (31) and VT-1598 (50), the para-position of a phenyl ring (like those for short tail) could have a week H-bond acceptor substitution, preferably oxygen (ether, carbonyl) and Nitrogen (NR2) that seems to form an H- bond with some residue in the active site such as S-378 or H-374 and H- 377 (in the case of VT-1598 and oteseconazole) [147,155,156], and also this H-bond acceptor group binds to another bulky aromatic group with possible steric and van de waals interactions in the active site. In the case of itraconazole-like drugs, a triazole-3-on group could enhance pharmacokinetics of a compound. This area in the active site has en­ ough space for further modification of structures to improve potency, physicochemical and pharmacokinetic of desired antifungal com­ pounds. 3. Concluding remarks and future directions The number of hosts for invasive fungal infections is growing fast due to an increase in circumstances that cause disturbed immune function. In addition, the resistant rate of infective fungal spp to drugs has improved with several known mechanisms like alteration in active site residue by point mutations in ERG11 as the responsible gen for CYP51, as well as ERG11 overexpression in fungal cells and MDR1 overexpression of efflux pump in fungi that may be concluded from insufficient drug therapy. Thus, common drugs are ineffective and in­ adequate to deal with this situation [157–160]. Although, four gen­ erations of azoles as most diverse antifungal drugs are well developed so far, the clinic has limited options for treatment. The options should contain a wide spectrum action, parenteral or oral antifungal drug with minimum drug-drug interaction and side-effects, effect on resistant fungal infections, and acceptable pharmacokinetics properties. This would never be achieved unless there are numerous studies by Table 5 fungal CYP51, Drug − Target Complexes in PDB Databank (rcsb.org) as Assessed in august 2019. Organism Ligand PDB code Resolution (A⁰) A.famigatus VNI 6CR2 2.38 A.famigatus VT-1598 5FRB 2.99 C. Albicans Itraconazole 5V5Z 2.9 C. Albicans Posaconazole 5FSA 2.86 C. Albicans VT-1161 5TZ1 2.0 C. Glabrata Itraconazole 5JLC 2.4 S. Cerevisiae Posaconazole 6E8Q 2.2 S. Cerevisiae Fluconazole 4WMZ 2.05 S. Cerevisiae VT-1161 5UL0 2.2 S. Cerevisiae Voriconazole 5HS1 2.1 S. Cerevisiae Lanosterol 4LXJ 1.9 S. Cerevisiae Itraconazole 5EQB 2.19 S. Cerevisiae (Y140F mutant) Itraconazole 4ZDY 2.02 S. Cerevisiae (Y140F mutant) Fluconazole 4ZDZ 2.3 S. Cerevisiae (Y140F mutant) Voriconazole 4ZE0 2.2 S. Cerevisiae (6464S mutant) Fluconazole 5ESY 2.15 S. Cerevisiae (G464S mutant) Itraconazole 5ESK 2.24 S. Cerevisiae (G73E mutant) Fluconazole 5ESF 2.25 S. Cerevisiae (G73E mutant) Itraconazole 5ESG 1.98 S. Cerevisiae (G73R mutant) Fluconazole 5ESE 2.2 S. Cerevisiae (G73W mutant) Itraconazole 5ESH 2.15 S. Cerevisiae (T3221 mutant) Itraconazole 5ESL 2.35 S. Cerevisiae (T3221 mutant) Fluconazole 5ESM 2.0 S. Cerevisiae (Y140Fmutant) Posaconazole 4ZE1 2.05 S. Cerevisiae (Y140H mutant) Itraconazole 4ZE2 2.3 S. Cerevisiae (Y140H mutant) Fluconazole 4ZE3 2.2 Fig. 15. Azole common structural model. M. Shafiei, et al. Bioorganic Chemistry 104 (2020) 104240 17
  • 18. medicinal chemistry scientist, the path started with fluconazole (24) discovery and is on road with the newest 4th generation VT-1598 (50) with improved characteristics. Albeit, in this way, azole is not the only solution to the problem and identification of new targets are important for efficient therapy. In this review, efforts were made to raise medic­ inal chemists’ awareness toward azole from their ancestor chlormida­ zole (1) to the most recent drug candidates in clinical trials and for the first time introduce them in four generations with different new structures. Moreover, hybrid antifungal agents as possible future gen­ eration which attracted high interest, were presented. The structure activity relationship according to valid and authentic information from SAR studies were also reported. Even though, in each study on the novel efficient structures the target-based information, based on new X-ray of CYP51 of wild or resistant types are unavoidable, we inform researchers of the recently uploaded information on the protein data bank (PDB, rcsb.org) for discovery and development of new members in the azole family. Declaration of Competing Interest The authors declared that there is no conflict of interest. Acknowledgements This work was supported by a grant from the research council of Tehran University of Medical Sciences (Grant no: 98-02-33-42922). Consent for publication Not applicable. References [1] G.D. Brown, D.W. Denning, N.A.R. Gow, S.M. Levitz, M.G. Netea, T.C. White, Supplementary Materials for Hidden Killers : Human Fungal Infections 13 (2012) 4–6, https://doi.org/10.1126/scitranslmed.3004404. [2] M.K. Kathiravan, A.B. Salake, A.S. Chothe, P.B. Dudhe, R.P. Watode, M.S. Mukta, S. Gadhwe, The biology and chemistry of antifungal agents: A review, Bioorganic Med. Chem. 20 (2012) 5678–5698, https://doi.org/10.1016/j.bmc.2012.04.045. [3] N. Thamban Chandrika, E.K. Dennis, S.K. Shrestha, H.X. Ngo, K.D. Green, S. Kwiatkowski, A.G. Deaciuc, L.P. Dwoskin, D.S. Watt, S. Garneau-Tsodikova, N,N′- diaryl-bishydrazones in a biphenyl platform: Broad spectrum antifungal agents, Eur. J. Med. Chem. (2019) 273–281. doi:10.1016/j.ejmech.2018.12.042. [4] J.R. Perfect, The antifungal pipeline: A reality check, Nat. Rev. Drug Discov. 16 (2017) 603–616, https://doi.org/10.1038/nrd.2017.46. [5] M. Ikeh, Y. Ahmed, J. Quinn, Phosphate Acquisition and Virulence in Human Fungal Pathogens, Microorganisms. 5 (2017) 48, https://doi.org/10.3390/ microorganisms5030048. [6] G.D. Brown, D.W. Denning, S.M. Levitz, Tackling human fungal infections, Science (80-.). 336 (2012) 647. doi:10.1126/science.1222236. [7] T. Roemer, D.J. Krysan, Unmet Clinical Needs, and New Approaches, Cold Spring Harb Perspect Med. (2014) 1–14, https://doi.org/10.1101/cshperspect.a019703. [8] J. Perlroth, B. Choi, B. Spellberg, Nosocomial fungal infections: Epidemiology, diagnosis, and treatment, Med. Mycol. 45 (2007) 321–346, https://doi.org/10. 1080/13693780701218689. [9] R. Calderone, N. Sun, F. Gay-Andrieu, W. Groutas, P. Weerawarna, S. Prasad, D. Alex, D. Li, Antifungal drug discovery: The process and outcomes, Future Microbiol. 9 (2014) 791–805, https://doi.org/10.2217/fmb.14.32. [10] M.E. Rodrigues, S. Silva, J. Azeredo, M. Henriques, Novel strategies to fight Candida species infection, Crit. Rev. Microbiol. 42 (2016) 594–606, https://doi. org/10.3109/1040841X.2014.974500. [11] M.A. Pfaller, D.J. Diekema, Epidemiology of invasive candidiasis: A persistent public health problem, Clin. Microbiol. Rev. 20 (2007) 133–163, https://doi.org/ 10.1128/CMR.00029-06. [12] V. Onnis, A. De Logu, M.T. Cocco, R. Fadda, R. Meleddu, C. Congiu, 2- Acylhydrazino-5-arylpyrrole derivatives: Synthesis and antifungal activity eva­ luation, Eur. J. Med. Chem. 44 (2009) 1288–1295, https://doi.org/10.1016/j. ejmech.2008.08.003. [13] A. Wald, W. Leisenring, J. van Burik, R.A. Bowden, Epidemiology of Aspergillus Infections in a Large Cohort of Patients Undergoing Bone Marrow Transplantation, J. Infect. Dis. 175 (1997) 1459–1466, https://doi.org/10.1086/516480. [14] D.A. Enoch, H. Yang, S.H. Aliyu, C. Micallef, Human Fungal Pathogen Identification (2017), https://doi.org/10.1007/978-1-4939-6515-1. [15] R.A. Fromtling, Overview of medically important antifungal azole derivatives, Clin. Microbiol. Rev. 1 (1988) 187–217, https://doi.org/10.1128/CMR.1.2.187. [16] J.P. Utz, Chemotherapy for the Systemic Mycoses: The Prelude to Ketoconazole, Clin. Infect. Dis. 2 (2011) 625–632, https://doi.org/10.1093/clinids/2.4.625. [17] A. Butts, D.J. Krysan, Antifungal Drug Discovery: Something Old and Something New, PLoS Pathog. 8 (2012) 9–11, https://doi.org/10.1371/journal.ppat. 1002870. [18] A.H. Aly, A. Debbab, P. Proksch, Fifty years of drug discovery from fungi, Fungal Divers. 50 (2011) 3–19, https://doi.org/10.1007/s13225-011-0116-y. [19] T.K. Mazu, B.A. Bricker, H. Flores-Rozas, S.Y. Ablordeppey, The Mechanistic Targets of Antifungal Agents: An Overview (2016), https://doi.org/10.2174/ 1389557516666160118112103. [20] D.S. Wishart, Y.D. Feunang, A.C. Guo, E.J. Lo, A. Marcu, J.R. Grant, T. Sajed, D. Johnson, C. Li, Z. Sayeeda, N. Assempour, I. Iynkkaran, Y. Liu, A. MacIejewski, N. Gale, A. Wilson, L. Chin, R. Cummings, Di. Le, A. Pon, C. Knox, M. Wilson, DrugBank 5.0: A major update to the DrugBank database for 2018, Nucleic Acids Res. 46 (2018) D1074–D1082. doi:10.1093/nar/gkx1037. [21] H. Su, L. Han, X. Huang, Potential targets for the development of new antifungal drugs, J. Antibiot. (Tokyo) 71 (2018) 978–991, https://doi.org/10.1038/s41429- 018-0100-9. [22] N. Liu, J. Tu, G. Dong, Y. Wang, C. Sheng, Emerging New Targets for the Treatment of Resistant Fungal Infections, J. Med. Chem. 61 (2018) 5484–5511, https://doi.org/10.1021/acs.jmedchem.7b01413. [23] C. Sheng, W. Zhang, New Lead Structures in Antifungal Drug Discovery, Curr. Med. Chem. 18 (2011) 733–766, https://doi.org/10.2174/092986711794480113. [24] L.R. Peyton, S. Gallagher, M. Hashemzadeh, Triazole antifungals: A review, Drugs of Today. 51 (2015) 705–718, https://doi.org/10.1358/dot.2015.51.12.2421058. [25] M.A. Hossain, M.A. Ghannoum, New investigational antifungal agents for treating invasive fungal infections, Expert Opin. Invest. Drugs 9 (2005) 1797–1813, https://doi.org/10.1517/13543784.9.8.1797. [26] M.C.M. Marı́a Victoria Castelli, Estefanı́a Butassi, F.V.& S.A.Z. Laura A Svetaz, Novel antifungal agents: a patent review (2011 - present), Expert Opin. Ther. Pat. 27 (2014) 415–426. doi:10.1517/13543776.2014.876993. [27] J.A. Paiva, J.M. Pereira, New antifungal antibiotics, Curr. Opin. Infect. Dis. 26 (2013) 168–174, https://doi.org/10.1097/QCO.0b013e32835ebcb7. [28] D.W. Woolley, Some biological effects produced by benzimidazole and their re­ versal by purines, J. Biol. Chem. (1944) 225–233. [29] H. van den Bossche, Biochemical effects of miconazole on fungi—I, Biochem. Pharmacol. 23 (2002) 887–899, https://doi.org/10.1016/0006-2952(74)90220-2. [30] V.H.P.R. Seeliger, Pilzhemmende Wirkung eines neuen Benzimidazol-Derivates, Mycoses 1 (1958) 162–171, https://doi.org/10.1111/j.1439-0507.1958. tb04016.x. [31] J.A. Maertens, History of the development of azole derivatives, Clin. Microbiol. Infect. 10 (2004) 1–10, https://doi.org/10.1111/j.1470-9465.2004.00841.x. [32] J.Y. Choi, L.M. Podust, W.R. Roush, Drug strategies targeting CYP51 in neglected tropical diseases, Chem. Rev. 114 (2014) 11242–11271, https://doi.org/10.1021/ cr5003134. [33] D. Allen, D. Wilson, R. Drew, J. Perfect, Azole antifungals: 35 years of invasive fungal infection management, Expert Rev. Anti. Infect. Ther. 13 (2015) 787–798, https://doi.org/10.1586/14787210.2015.1032939. [34] V. Fainstein, G.P. Bodey, Cardiorespiratory Toxicity Due to Miconazole, Ann. Intern. Med. 93 (2013) 432, https://doi.org/10.7326/0003-4819-93-3-432. [35] P.R. Sawyer, R.N. Brogden, T.M. Speight, G.S. Avery, clotrimazole: A Review of its Antifungal Activity and Therapeutic Efficacy, Drugs. 9 (1975) 424–447, https:// doi.org/10.2165/00003495-197509060-00003. [36] D. Khanna, S. Bharti, Luliconazole for the treatment of fungal infections: an evi­ dence-based review, Core Evid. (2014) 113, https://doi.org/10.2147/ce.s49629. [37] J. Heeres, L. Backx, J. Mostmans, J. Van Cutsem, Antimycotic imidazoles. Part 4. Synthesis and antifungal activity of ketoconazole, a new potent orally active broad-spectrum antifungal agent, J. Med. Chem. 22 (1979) 1003–1005. [38] E. Drouhet, B. Dupont, Chronic Mucocutaneous Candidosis and Other Superficial and Systemic Mycoses Successfully Treated with Ketoconazole, Clin. Infect. Dis. 2 (2011) 606–619, https://doi.org/10.1093/clinids/2.4.606. [39] nizoral fda, (n.d.). www.fda.gov/Drugs/DrugSafety/ucm362415.htm. [40] K. Richardson, The discovery and profile of fluconazole, J. Chemother. 2 (1990) 51–54, https://doi.org/10.1080/1120009X.1990.11738981. [41] R. Cha, J.D. Sobel, Fluconazole for the treatment of candidiasis: 15 years experi­ ence, Expert Rev. Anti. Infect. Ther. 2 (2004) 357–366, https://doi.org/10.1586/ 14787210.2.3.357. [42] P.F. Troke, R.J. Andrews, G.W. Pye, K. Richardson, Fluconazole and other azoles: Translation of in vitro activity to in vivo and clinical efficacy, Rev. Infect. Dis. 12 (1990) S276–S280, https://doi.org/10.1093/clinids/12.Supplement_3.S276. [43] R.O. Darouiche, D. Abi-Said, D.P. Kontoyiannis, L.O. Gentry, E.J. Anaissie, T. Williams, G.P. Bodey, J. Mera, C.L. Karl, O. Uzun, Management of Invasive Candidal Infections: Results of a Prospective, Randomized, Multicenter Study of Fluconazole Versus Amphotericin B and Review of the Literature, Clin. Infect. Dis. 23 (2011) 964–972, https://doi.org/10.1093/clinids/23.5.964. [44] A. Bentley, M. Butters, S.P. Green, W.J. Learmonth, J.A. MacRae, M.C. Morland, G. O’Connor, J. Skuse, The discovery and process development of a commercial route to the water soluble prodrug, fosfluconazole, Org. Process Res. Dev. 6 (2002) 109–112, https://doi.org/10.1021/op010064+. [45] S. Sobue, K. Tan, G. Layton, M. Eve, J.B. Sanderson, Pharmacokinetics of fos­ fluconazole and fluconazole following multiple intravenous administration of fosfluconazole in healthy male volunteers, Br. J. Clin. Pharmacol. 58 (2004) 20–25, https://doi.org/10.1111/j.1365-2125.2004.02107.x. [46] D. Abraham, Chemotherapeutic Agents, in: Burger’s Med. Chem. Drug Discov., John Sons, Inc, 2003: pp. 881–910. [47] E.J. Corey, M. Chaykovsky, Dimethylsulfonium Methylide, A Reagent for Selective M. Shafiei, et al. Bioorganic Chemistry 104 (2020) 104240 18
  • 19. Oxirane Synthesis from Aldehydes and Ketones, J. Am. Chem. Soc. 84 (1962) 3782–3783, https://doi.org/10.1021/ja00878a046. [48] E.J. Corey, M. Chaykovsky, Methylsulfinyl Carbanion (CH 3 -SO-CH 2). Formation and Applications to Organic Synthesis, J. Am. Chem. Soc. 87 (1965) 1345–1353, https://doi.org/10.1021/ja01084a033. [49] E.J. Corey, M. Chaykovsky, Formation and Photochemical Rearrangement of β’- Ketosulfoxonium Ylides, J. Am. Chem. Soc. 86 (1964) 1640–1641, https://doi. org/10.1021/ja01062a040. [50] J.J. Li, Corey-Chaykovsky epoxidation, Name React. (2002), https://doi.org/10. 1007/978-3-662-04835-1_64 77 77. [51] M.J. Dolton, A.J. McLachlan, Optimizing azole antifungal therapy in the prophy­ laxis and treatment of fungal infections, Curr. Opin. Infect. Dis. 27 (2014) 493–500, https://doi.org/10.1097/QCO.0000000000000103. [52] M.W. Pound, M.L. Townsend, V. Dimond, D. Wilson, R.H. Drew, Overview of treatment options for invasive fungal infections, Med. Mycol. 49 (2011) 561–580, https://doi.org/10.3109/13693786.2011.560197. [53] C. Lass-Flörl, Triazole antifungal agents in invasive fungal infections: A com­ parative review, Drugs. 71 (2011) 2405–2419, https://doi.org/10.2165/ 11596540-000000000-00000. [54] S.W. Chapman, W.E. Dismukes, L.A. Proia, R.W. Bradsher, P.G. Pappas, M.G. Threlkeld, C.A. Kauffman, Clinical Practice Guidelines for the Management of Blastomycosis: 2008 Update by the Infectious Diseases Society of America, Clin. Infect. Dis. 46 (2008) 1801–1812. doi:10.1086/588300. [55] D.A. Stevens, J.Y. Lee, Analysis Of Compassionate Use Itraconazole Therapy For Invasive Aspergillosis By The Niaid Mycoses Study Group Criteria, Arch. Intern. Med. 157 (1997) 1857–1862. [56] L. Willems, R. Van der Geest, K. de Beule, Itraconazole oral solution and in­ travenous formulations: a review of pharmacokinetics and pharmacodynamics, J. Clin. Pharm. Ther. 26 (2001) 159–169 https://www.patientslikeme.com. [57] T.K. Daneshmend, D.W. Warnock, Clinical Pharmacokinetics of Ketoconazole, Clin. Pharmacokinet. 14 (1988) 13–34, https://doi.org/10.2165/00003088- 198814010-00002. [58] L. Jeu, F.J. Piacenti, A.G. Lyakhovetskiy, H.B. Fung, New bugs Voriconazole, New York. (2003). [59] F. Ezzet, D. Wexler, R. Courtney, G. Krishna, J. Lim, M. Laughlin, Oral Bioavailability of Posaconazole in Fasted Healthy Subjects, Clin. Pharmacokinet. 44 (2005) 211–220, https://doi.org/10.2165/00003088-200544020-00006. [60] R. Bellmann, P. Smuszkiewicz, Pharmacokinetics of antifungal drugs: practical implications for optimized treatment of patients, Infection 45 (2017) 737–779, https://doi.org/10.1007/s15010-017-1042-z. [61] S.R. Lipner, R.K. Scher, Efinaconazole in the treatment of onychomycosis, Infect. Drug Resist. 8 (2015) 163–172, https://doi.org/10.2147/IDR.S69596. [62] A.C. Pasqualotto, K.O. Thiele, L.Z. Goldani, Novel triazole antifungal drugs: focus on isavuconazole, ravuconazole and albaconazole, Curr. Opin. Invest. Drugs 11 (2010) 165–174 http://www.ncbi.nlm.nih.gov/pubmed/20112166. [63] A.J. Dietz, J.C. Barnard, K. van Rossem, A randomized, double-blind, multiple- dose, placebo-controlled, dose escalation study with a 3-cohort parallel group design to investigate the tolerability and pharmacokinetics of albaconazole in healthy subjects, Clin. Pharmacol. Drug Dev. 3 (2014) 25–33, https://doi.org/10. 1002/cpdd.72. [64] C. Lin, H. Kim, E. Radwanski, M. Affrime, M. Brannan, M.N. Cayen, Pharmacokinetics and metabolism of genaconazole, a potent antifungal drug, in men, Antimicrob. Agents Chemother. 40 (1996) 92–96, https://doi.org/10.1128/ AAC.40.1.92. [65] N. Sun, Y. Xie, C. Sheng, Y. Cao, W. Zhang, H. Chen, G. Fan, In vivo pharmaco­ kinetics and in vitro antifungal activity of iodiconazole, a new triazole, determined by microdialysis sampling, Int. J. Antimicrob. Agents 41 (2013) 229–235, https:// doi.org/10.1016/j.ijantimicag.2012.10.020. [66] Y. Kamai, M. Kubota, T. Fukuoka, Y. Kamai, N. Maeda, T. Hosokawa, T. Shibayama, K. Uchida, H. Yamaguchi, S. Kuwahara, Efficacy of CS-758, a novel triazole, against experimental fluconazole-resistant oropharyngeal candidiasis in mice, Antimicrob. Agents Chemother. 47 (2003) 601–606, https://doi.org/10. 1128/AAC.47.2.601-606.2003. [67] C. Girmenia, E. Finolezzi, New-generation triazole antifungal drugs: review of the Phase II and III trials, Clin. Investig. (Lond) 1 (2011) 1577–1594, https://doi.org/ 10.4155/cli.11.137. [68] L.B. Johnson, C.A. Kuffman, Voriconazole: A new triazole antifungal agent, Rev. ANTI-INFECTIVE AGENTS. 36 (2003) 630 637 doi:1058-4838/2003/3605- 0014$15.00. [69] F. Sabatelli, R. Patel, P.A. Mann, C.A. Mendrick, C.C. Norris, R. Hare, D. Loebenberg, T.A. Black, P.M. McNicholas, In vitro activities of posaconazole, fluconazole, itraconazole, voriconazole, and amphotericin B against a large col­ lection of clinically important molds and yeasts, Antimicrob. Agents Chemother. 50 (2006) 2009–2015, https://doi.org/10.1128/AAC.00163-06. [70] F. Bennett, A.K. Saksena, R.G. Lovey, Y.T. Liu, N.M. Patel, P. Pinto, R. Pike, E. Jao, V.M. Girijavallabhan, A.K. Ganguly, D. Loebenberg, H. Wang, A. Cacciapuoti, E. Moss, F. Menzel, R.S. Hare, A. Nomeir, Hydroxylated analogues of the orally active broad spectrum antifungal, Sch 51048 (1), and the discovery of posaco­ nazole [Sch 56592; 2 or (S, S)-5], Bioorganic Med. Chem. Lett. 16 (2006) 186–190, https://doi.org/10.1016/j.bmcl.2005.09.031. [71] A. Nomeir, B.N. Pramanik, L. Heimark, F. Bennett, J. Veals, P. Bartner, M. Hilbert, A. Saksena, P. McNamara, Posaconazole (Noxafil, SCH 56592), a new azole an­ tifungal drug, was a discovery based on the isolation and mass spectral char­ acterization of a circulating metabolite of an earlier lead (SCH 51048), J. Mass Spectrom. 43 (2008) 509–517, https://doi.org/10.1002/jms.1341. [72] I.I. Raad, J.R. Graybill, A.B. Bustamante, O.A. Cornely, V. Gaona-Flores, C. Afif, D.R. Graham, R.N. Greenberg, S. Hadley, A. Langston, R. Negroni, J.R. Perfect, P. Pitisuttithum, A. Restrepo, G. Schiller, L. Pedicone, A.J. Ullmann, Safety of Long-Term Oral Posaconazole Use in the Treatment of Refractory Invasive Fungal Infections, Clin. Infect. Dis. 42 (2006) 1726–1734, https://doi.org/10.1086/ 504328. [73] O.A. Cornely, J. Maertens, D.J. Winston, J. Perfect, A.J. Ullmann, T.J. Walsh, D. Helfgott, J. Holowiecki, D. Stockelberg, Y.-T. Goh, M. Petrini, C. Hardalo, R. Suresh, D. Angulo-Gonzalez, Posaconazole vs. Fluconazole or Itraconazole Prophylaxis in Patients with Neutropenia, N. Engl. J. Med. 356 (2007) 348–359, https://doi.org/10.1056/nejmoa061094. [74] M. Andrew J. Ullmann, M.D., Jeffrey H. Lipton, M.D., David H. Vesole, M.D., Ph. D., Pranatharthi Chandrasekar, M.D., Amelia Langston, M.D., Stefano R. Tarantolo, M.D., Hildegard Greinix, M.D., Wellington Morais de Azevedo, M.D., Ph.D., Vijay Reddy, M.D., Navd, Posaconazole or Fluconazole for Prophylaxis in Severe Graft-versus-Host Disease, N. Engl. J. Med. 356 (2007) 335–347. [75] T.J. Walsh, I. Raad, T.F. Patterson, P. Chandrasekar, G.R. Donowitz, R. Graybill, R.E. Greene, R. Hachem, S. Hadley, R. Herbrecht, A. Langston, A. Louie, P. Ribaud, B.H. Segal, D.A. Stevens, J.H. van Burik, C.S. White, G. Corcoran, J. Gogate, G. Krishna, L. Pedicone, C. Hardalo, J.R. Perfect, Treatment of Invasive Aspergillosis with Posaconazole in Patients Who Are Refractory to or Intolerant of Conventional Therapy: An Externally Controlled Trial, Clin. Infect. Dis. 44 (2007) 2–12, https://doi.org/10.1086/508774. [76] T. Patel, S. Dhillon, Efinaconazole: First global approval, Drugs. 73 (2013) 1977–1983, https://doi.org/10.1007/s40265-013-0152-x. [77] A. Schmitt-hoffmann, B. Roos, M. Heep, M. Schleimer, E. Weidekamm, T. Brown, M. Roehrle, C. Beglinger, Single-Ascending-Dose Pharmacokinetics and Safety of the Novel Broad-Spectrum Antifungal Triazole BAL4815 after Intravenous Infusions (50, 100, and 200 Milligrams) and Oral Administrations (100, 200, and 400 Milligrams) of Its Prodrug, BAL8557, in Healthy, Antimicrob. Agents Chemother. 50 (2006) 279–285, https://doi.org/10.1128/AAC.50.1.279-285. 2006. [78] boyd 83, (n.d.). https://www.aspergillus.org.uk/content/bal-4815bal-8557-anti­ fungal-agent. [79] S. Seyedmousavi, P.E. Verweij, J.W. Mouton, Isavuconazole, a broad-spectrum triazole for the treatment of systemic fungal diseases, Expert Rev. Anti. Infect. Ther. 13 (2015) 9–27, https://doi.org/10.1586/14787210.2015.990382. [80] N.N. Pettit, P.L. Carver, Isavuconazole: A New Option for the Management of Invasive Fungal Infections, Ann. Pharmacother. 49 (2015) 825–842, https://doi. org/10.1177/1060028015581679. [81] H. Yamaguchi, Potential of Ravuconazole and its Prodrugs as the New OralTherapeutics for Onychomycosis, Med. Mycol. J. 57 (2016) E93–E110, https://doi.org/10.3314/mmj.16-00006. [82] J. Capilla, M. Ortoneda, F.J. Pastor, J. Guarro, In vitro antifungal activities of the new triazole UR-9825 against clinically important filamentous fungi, Antimicrob. Agents Chemother. 45 (2001) 2635–2637, https://doi.org/10.1128/AAC.45.9. 2635-2637.2001. [83] clinical trial, (n.d.). https://clinicaltrials.gov. [84] Y.N. Cheng, Z.H. Jiang, L.S. Sun, Z.Y. Su, M.M. Zhang, H.L. Li, Synthesis of 1, 2, 4- triazole benzoyl arylamine derivatives and their high antifungal activities, Eur. J. Med. Chem. 200 (2020) 112463, https://doi.org/10.1016/j.ejmech.2020.112463. [85] Z. Ding, T. Ni, F. Xie, Y. Hao, S. Yu, X. Chai, Y. Jin, T. Wang, Y. Jiang, D. Zhang, Design, synthesis, and structure-activity relationship studies of novel triazole agents with strong antifungal activity against Aspergillus fumigatus, Bioorganic Med. Chem. Lett. 30 (2020) 126951, https://doi.org/10.1016/j.bmcl.2020. 126951. [86] F. Xie, T. Ni, Z. Ding, Y. Hao, R. Wang, R. Wang, T. Wang, X. Chai, S. Yu, Y. Jin, Y. Jiang, D. Zhang, Design, synthesis, and in vitro evaluation of novel triazole analogues featuring isoxazole moieties as antifungal agents, Bioorg. Chem. 101 (2020) 103982, https://doi.org/10.1016/j.bioorg.2020.103982. [87] İ.S. Doğan, S. Saraç, S. Sari, D. Kart, Ş. Eşsiz Gökhan, İ. Vural, S. Dalkara, New azole derivatives showing antimicrobial effects and their mechanism of antifungal activity by molecular modeling studies, Eur. J. Med. Chem. 130 (2017) 124–138, https://doi.org/10.1016/j.ejmech.2017.02.035. [88] V.W. Rabelo, T.F. Santos, L. Terra, M.V. Santana, H.C. Castro, C.R. Rodrigues, P.A. Abreu, Targeting CYP51 for drug design by the contributions of molecular modeling, Fundam. Clin. Pharmacol. 31 (2017) 37–53, https://doi.org/10.1111/ fcp.12230. [89] Y. Zhang, V.K.R. Tangadanchu, R.R.Y. Bheemanaboina, Y. Cheng, C.H. Zhou, Novel carbazole-triazole conjugates as DNA-targeting membrane active potentia­ tors against clinical isolated fungi, Eur. J. Med. Chem. 155 (2018) 579–589, https://doi.org/10.1016/j.ejmech.2018.06.022. [90] Z. Jiang, J. Gu, C. Wang, S. Wang, N. Liu, Y. Jiang, G. Dong, Y. Wang, Y. Liu, J. Yao, Z. Miao, W. Zhang, C. Sheng, Design, synthesis and antifungal activity of novel triazole derivatives containing substituted 1,2,3-triazole-piperdine side chains, Eur. J. Med. Chem. 82 (2014) 490–497, https://doi.org/10.1016/j.ejmech. 2014.05.079. [91] S.K. Shrestha, A. Garzan, S. Garneau-Tsodikova, Novel alkylated azoles as potent antifungals, Eur. J. Med. Chem. 133 (2017) 309–318, https://doi.org/10.1016/j. ejmech.2017.03.075. [92] A. Qian, Y. Zheng, R. Wang, J. Wei, Y. Cui, X. Cao, Y. Yang, Design, synthesis, and structure-activity relationship studies of novel tetrazole antifungal agents with potent activity, broad antifungal spectrum and high selectivity, Bioorganic Med. Chem. Lett. 28 (2018) 344–350, https://doi.org/10.1016/j.bmcl.2017.12.040. [93] P.D. Patel, M.R. Patel, B. Kocsis, E. Kocsis, S.M. Graham, A.R. Warren, S.M. Nicholson, B. Billack, F.R. Fronczek, T.T. Talele, Design, synthesis and de­ termination of antifungal activity of 5(6)-substituted benzotriazoles, Eur. J. Med. M. Shafiei, et al. Bioorganic Chemistry 104 (2020) 104240 19
  • 20. Chem. 45 (2010) 2214–2222, https://doi.org/10.1016/j.ejmech.2010.01.062. [94] W. Wang, S. Wang, Y. Liu, G. Dong, Y. Cao, Z. Miao, J. Yao, W. Zhang, C. Sheng, Novel conformationally restricted triazole derivatives with potent antifungal ac­ tivity, Eur. J. Med. Chem. 45 (2010) 6020–6026, https://doi.org/10.1016/j. ejmech.2010.09.070. [95] Y. Zhang, G.L.V. Damu, S.F. Cui, J.L. Mi, V.K.R. Tangadanchu, C.H. Zhou, Discovery of potential antifungal triazoles: Design, synthesis, biological evalua­ tion, and preliminary antifungal mechanism exploration, Medchemcomm. 8 (2017) 1631–1639, https://doi.org/10.1039/c7md00112f. [96] H. Tang, J. Wu, W. Zhang, L. Zhao, Y.H. Zhang, C.W. Shen, Design, synthesis and biological evaluation of novel non-azole derivatives as potential antifungal agents, Chinese Chem. Lett. 26 (2015) 1161–1164, https://doi.org/10.1016/j.cclet.2015. 04.030. [97] N.P. Wiederhold, L.K. Najvar, E.P. Garvey, S.R. Brand, X. Xu, E.A. Ottinger, A. Alimardanov, J. Cradock, M. Behnke, W.J. Hoekstra, R.J. Schotzinger, R. Jaramillo, M. Olivo, W.R. Kirkpatrick, T.F. Patterson, The fungal cyp51 in­ hibitor vt-1129 is efficacious in an experimental model of cryptococcal meningitis, Antimicrob. Agents Chemother. 62 (2018) 1–10, https://doi.org/10.1128/AAC. 01071-1. [98] S.R. Brand, T.P. Degenhardt, K. Person, J.D. Sobel, P. Nyirjesy, R.J. Schotzinger, A. Tavakkol, A phase 2, randomized, double-blind, placebo-controlled, dose-ran­ ging study to evaluate the efficacy and safety of orally administered VT-1161 in the treatment of recurrent vulvovaginal candidiasis, Am. J. Obstet. Gynecol. 624 (e1–624) (2018) e9, https://doi.org/10.1016/j.ajog.2018.03.001. [99] W.J. Hoekstra, E.P. Garvey, W.R. Moore, S.W. Rafferty, C.M. Yates, R.J. Schotzinger, Design and optimization of highly-selective fungal CYP51 in­ hibitors, Bioorganic Med. Chem. Lett. 24 (2014) 3455–3458, https://doi.org/10. 1016/j.bmcl.2014.05.068. [100] C.M. Yates, E.P. Garvey, S.R. Shaver, R.J. Schotzinger, W.J. Hoekstra, Design and optimization of highly-selective, broad spectrum fungal CYP51 inhibitors, Bioorganic Med. Chem. Lett. 27 (2017) 3243–3248, https://doi.org/10.1016/j. bmcl.2017.06.037. [101] N.P. Wiederhold, H.P. Patterson, B.H. Tran, C.M. Yates, R.J. Schotzinger, E.P. Garvey, Fungal-specific Cyp51 inhibitor VT-1598 demonstrates in vitro ac­ tivity against Candida and Cryptococcus species, endemic fungi, including Coccidioides species, Aspergillus species and Rhizopus arrhizus, J. Antimicrob. Chemother. 73 (2018) 404–408, https://doi.org/10.1093/jac/dkx410. [102] A.G.S. Warrilow, J.E. Parker, C.L. Price, W.D. Nes, E.P. Garvey, W.J. Hoekstra, R.J. Schotzinger, D.E. Kelly, S.L. Kelly, The investigational drug VT-1129 is a highly potent inhibitor of Cryptococcus species CYP51 but only weakly inhibits the human enzyme, Antimicrob. Agents Chemother. 60 (2016) 4530–4538, https:// doi.org/10.1128/AAC.00349-16. [103] S. Brand, T. Degenhardt, P. Nyirjesy, J. Sobel, C. Handelsman, K. Person, R. Schotzinger, A. Tavakkol, 15: To evaluate the efficacy and safety of VT-1161, a potent, highly selective inhibitor of fungal CYP51, in treating women with a documented history of recurrent vulvovaginal candidiasis (RVVC), Am. J. Obstet. Gynecol. 215 (2016) S821, https://doi.org/10.1016/j.ajog.2016.09.016. [104] S.Q. Wang, Y.F. Wang, Z. Xu, Tetrazole hybrids and their antifungal activities, Eur. J. Med. Chem. (2019) 225–234, https://doi.org/10.1016/j.ejmech.2019.03.023. [105] N.P. Wiederhold, The antifungal arsenal: alternative drugs and future targets, Int. J. Antimicrob. Agents 51 (2018) 333–339, https://doi.org/10.1016/j.ijantimicag. 2017.09.002. [106] N.P. Wiederhold, L.K. Najvar, R. Jaramillo, M. Olivo, G. Catano, T.F. Patterson, L.F. Shubitz, H.T. Trinh, C.M. Yates, R.J. Schotzinger, E.P. Garvey, The novel fungal Cyp51 inhibitor VT-1598 is efficacious in experimental models of central nervous system coccidioidomycosis caused by Coccidioides posadasii and Coccidioides immitis, Antimicrob. Agents Chemother. 62 (2018) 1–7, https://doi. org/10.1128/AAC.02258-17. [107] N.P. Wiederhold, S.R. Lockhart, L.K. Najvar, E.L. Berkow, R. Jaramillo, M. Olivo, E.P. Garvey, C.M. Yates, R.J. Schotzinger, G. Catano, T.F. Patterson, The Fungal Cyp51-Specific Inhibitor VT-1598 Demonstrates In Vitro and In Vivo Activity against Candida auris, Antimicrob. Agents Chemother. (2019), https://doi.org/10. 1128/AAC.02233-18. [108] B. Meunier, Hybrid Molecules with a Dual Mode of Action : Dream or Reality ? †, Acc. Chem. Res. 41 (2008) 69–77. [109] F. Terms, An overview of molecular hybrids in drug discovery, Expert Opin. Drug Discov. (2016), https://doi.org/10.1517/17460441.2016.1135125. [110] H.R. Martins, J.D. De Souza, F. Marcelo, P.B. Victor, H.O.M. Jarbas, R.M. Verly, Glycotriazole - peptides derived from the peptide HSP1: synergistic effect of triazole and saccharide rings on the antifungal activity, Amino Acids 49 (2017) 1389–1400, https://doi.org/10.1007/s00726-017-2441-2. [111] S. Wu, Y. Zhang, X. He, X. Che, S. Wang, Y. Liu, Y. Jiang, N. Liu, G. Dong, J. Yao, Z. Miao, Y. Wang, From Antidiabetic to Antifungal : Discovery of Highly Potent Triazole – Thiazolidinedione Hybrids as Novel Antifungal Agents, ChemMedChem 9 (2014) 2639–2646, https://doi.org/10.1002/cmdc.201402320. [112] S. Emami, E. Ghobadi, S. Saednia, S.M. Hashemi, Current advances of triazole alcohols derived from fluconazole: Design, in vitro and in silico studies, Eur. J. Med. Chem. (2019) 173–194, https://doi.org/10.1016/j.ejmech.2019.03.020. [113] N. Belskaya, J. Subbotina, S. Lesogorova, Synthesis of 2 H -1, 2, 3-Triazoles, Top Heterocycl Chem. 1 (2015) 51–116, https://doi.org/10.1007/7081. [114] Y. Wang, K. Xu, G. Bai, L. Huang, Q. Wu, W. Pan, S. Yu, Synthesis and Antifungal Activity of Novel Triazole Compounds Containing Piperazine Moiety, Molecules 19 (2014) 11333–11340, https://doi.org/10.3390/molecules190811333. [115] N.D. Thanh, D.S. Hai, V.T. Ngoc Bich, P.T. Thu Hien, N.T. Ky Duyen, N.T. Mai, T.T. Dung, V.N. Toan, H.T. Kim Van, L.H. Dang, D.N. Toan, T.T. Thanh Van, Efficient click chemistry towards novel 1H-1,2,3-triazole-tethered 4H-chromene−D- glucose conjugates: Design, synthesis and evaluation of in vitro antibacterial, MRSA and antifungal activities, Eur. J. Med. Chem. (2019) 454–471. doi:10.1016/ j.ejmech.2019.01.060. [116] Y. Zou, Q. Zhao, J. Liao, H. Hu, S. Yu, X. Chai, M. Xu, Q. Wu, Bioorganic & Medicinal Chemistry Letters New triazole derivatives as antifungal agents : Synthesis via click reaction, in vitro evaluation and molecular docking studies, Bioorg. Med. Chem. Lett. 22 (2012) 2959–2962, https://doi.org/10.1016/j.bmcl. 2012.02.042. [117] N. Fu, S. Wang, Y. Zhang, C. Zhang, D. Yang, L. Weng, B. Zhao, L. Wang, Efficient click chemistry towards fatty acids containing 1,2,3-triazole: Design and synthesis as potential antifungal drugs for Candida albicans, Eur. J. Med. Chem. (2017), https://doi.org/10.1016/j.ejmech.2017.05.001. [118] V.S. Pore, N.G. Aher, P.K. Shukla, Design and synthesis of fluconazole / bile acid conjugate using click reaction, Tetrahedron 62 (2006) 11178–11186, https://doi. org/10.1016/j.tet.2006.09.021. [119] K. Bozorov, J. Zhao, H.A. Aisa, 1,2,3-Triazole-containing hybrids as leads in medicinal chemistry: A recent overview, Bioorganic Med. Chem. 27 (2019) 3511–3531, https://doi.org/10.1016/j.bmc.2019.07.005. [120] A. Gluszynska, Biological potential of carbazole derivatives, Eur. J. Med. Chem. 94 (2015) 405–426, https://doi.org/10.1016/j.ejmech.2015.02.059. [121] R. Elias, R.I. Benhamou, Q.Z. Jaber, O. Dorot, S.L. Zada, K. Oved, E. Pichinuk, M. Fridman, Antifungal activity, mode of action variability, and subcellular dis­ tribution of coumarin-based antifungal azoles, Eur. J. Med. Chem. 179 (2019) 779–790, https://doi.org/10.1016/j.ejmech.2019.07.003. [122] G. Han, N. Liu, C. Li, J. Tu, Z. Li, C. Sheng, Discovery of Novel Fungal Lanosterol 14 α -Demethylase (CYP51)/ Histone Deacetylase (HDAC) Dual Inhibitors to treat azole-resistant candidiasis, J. Med. Chem. Discov. Apr (2020). doi:10.1021/acs. jmedchem.0c00102. [123] X. Fang, D. Li, V. Kumar, R. Tangadanchu, L. Gopala, W. Gao, Novel potentially antifungal hybrids of 5-flucytosine and fluconazole : Design, synthesis and bioactive evaluation, Bioorg. Med. Chem. Lett. (2017), https://doi.org/10.1016/j. bmcl.2017.10.020. [124] T. Režen, N. Debeljak, D. Kordiš, D. Rozman, New aspects on lanosterol 14α-de­ methylase and cytochrome P450 evolution: Lanosterol/cycloartenol diversifica­ tion and lateral transfer, J. Mol. Evol. 59 (2004) 51–58, https://doi.org/10.1007/ s00239-004-2603-1. [125] E. Desmond, S. Gribaldo, Phylogenomics of Sterol Synthesis: Insights into the Origin, Evolution, and Diversity of a Key Eukaryotic Feature, Genome Biol. Evol. 1 (2009) 364–381, https://doi.org/10.1093/gbe/evp036. [126] M.V. Keniya, M. Sabherwal, R.K. Wilson, M.A. Woods, A.A. Sagatova, J.D.A. Tyndall, Crystal Structures of Full-Length Lanosterol 14α-Demethylases of Prominent Fungal Pathogens Candida albicans and Candida glabrata Provide Tools for Antifungal Discovery, Antimicrob. Agents Chemother. 62 (2018) e01134–e1218. [127] F. Hannemann, A. Bichet, K.M. Ewen, R. Bernhardt, Cytochrome P450 systems- biological variations of electron transport chains, Biochim. Biophys. Acta - Gen. Subj. 1770 (2007) 330–344, https://doi.org/10.1016/j.bbagen.2006.07.017. [128] T.Y. Hargrove, Z. Wawrzak, D.C. Lamb, F.P. Guengerich, G.I. Lepesheva, Structure-functional characterization of cytochrome P450 Sterol 14α-Demethylase (CYP51B) from aspergillus fumigatus and molecular basis for the development of antifungal drugs, J. Biol. Chem. 290 (2015) 23916–23934, https://doi.org/10. 1074/jbc.M115.677310. [129] B.C. Monk, A.A. Sagatova, P. Hosseini, Y.N. Ruma, R.K. Wilson, M.V. Keniya, Fungal Lanosterol 14α-demethylase: A target for next-generation antifungal de­ sign, Biochim. Biophys. Acta - Proteins Proteomics. (2019), https://doi.org/10. 1016/j.bbapap.2019.02.008. [130] J. Zhang, L. Li, Q. Lv, L. Yan, Y. Wang, Y. Jiang, The fungal CYP51s: Their func­ tions, structures, related drug resistance, and inhibitors, Front. Microbiol. 10 (2019), https://doi.org/10.3389/fmicb.2019.00691. [131] F.V. Galina. Lepesheva, Tatiana Y. Hargrove1, Girish Rachakonda, Zdzislaw Wawrzak, Sébastien Pomel, Sandrine Cojean, Pius N. Nde, W. David Nes, Charles W. Locuson, M. Wade Calcutt, Michael R. Waterman, J. Scott Daniels, Philippe M. Loiseau, VFV as a new effective CYP51 structure-derived drug candidate for Chagas disease and visceral leishmaniasis, J. Infect. Dis. April 15 (2015) 1–25. doi:10.1093/infdis/jiv228. [132] F. Villalta, M.C. Dobish, P.N. Nde, Y.Y. Kleshchenko, T.Y. Hargrove, C.A. Johnson, M.R. Waterman, J.N. Johnston, G.I. Lepesheva, VNI cures acute and chronic ex­ perimental chagas disease, J. Infect. Dis. 208 (2013) 504–511, https://doi.org/10. 1093/infdis/jit042. [133] G.I. Lepesheva, H.W. Park, T.Y. Hargrove, B. Vanhollebeke, Z. Wawrzak, J.M. Harp, M. Sundaramoorthy, W.D. Nes, E. Pays, M. Chaudhuri, F. Villalta, M.R. Waterman, Crystal structures of Trypanosoma brucei sterol 14α-demethylase and implications for selective treatment of human infections, J. Biol. Chem. 285 (2010) 1773–1780, https://doi.org/10.1074/jbc.M109.067470. [134] T.Y. Hargrove, L. Friggeri, Z. Wawrzak, A. Qi, W.J. Hoekstra, R.J. Schotzinger, J.D. York, F. Peter Guengerich, G.I. Lepesheva, Structural analyses of Candida albicans sterol 14α-demethylase complexed with azole drugs address the mole­ cular basis of azole-mediated inhibition of fungal sterol biosynthesis, J. Biol. Chem. 292 (2017) 6728–6743, https://doi.org/10.1074/jbc.M117.778308. [135] G.I. Lepesheva, M.R. Waterman, Structural basis for conservation in the CYP51 family, Biochim. Biophys. Acta - Proteins Proteomics. 2011 (1814) 88–93, https:// doi.org/10.1016/j.bbapap.2010.06.006. [136] A.A. Sagatova, M.V. Keniya, R.K. Wilson, B.C. Monk, J.D.A. Tyndall, Structural insights into binding of the antifungal drug fluconazole to Saccharomyces cere­ visiae lanosterol 14α-demethylase, Antimicrob. Agents Chemother. 59 (2015) 4982–4989, https://doi.org/10.1128/AAC.00925-15. M. Shafiei, et al. Bioorganic Chemistry 104 (2020) 104240 20
  • 21. [137] C. Sheng, S. Chen, H. Ji, G. Dong, X. Che, W. Wang, Z. Miao, J. Yao, J. Lü, W. Guo, W. Zhang, Evolutionary trace analysis of CYP51 family: Implication for site-di­ rected mutagenesis and novel antifungal drug design, J. Mol. Model. 16 (2010) 279–284, https://doi.org/10.1007/s00894-009-0527-9. [138] B.C. Monk, M.V. Keniya, M. Sabherwal, R.K. Wilson, D.O. Graham, H.F. Hassan, D. Chen, J.D.A. Tyndall, Azole resistance reduces susceptibility to the tetrazole antifungal VT-1161, Antimicrob. Agents Chemother. 63 (2019) 1–19, https://doi. org/10.1128/AAC.02114-18. [139] 5ul0 116, (n.d.). https://www.rcsb.org/structure/5ul0. [140] B.C. Monk, T.M. Tomasiak, M.V. Keniya, F.U. Huschmann, J.D.A. Tyndall, J.D. O’Connell, R.D. Cannon, J.G. McDonald, A. Rodriguez, J.S. Finer-Moore, R.M. Stroud, Architecture of a single membrane spanning cytochrome P450 sug­ gests constraints that orient the catalytic domain relative to a bilayer, Proc. Natl. Acad. Sci. 111 (2014) 3865–3870, https://doi.org/10.1073/pnas.1324245111. [141] L. Friggeri, T.Y. Hargrove, Z. Wawrzak, A.L. Blobaum, G. Rachakonda, C.W. Lindsley, F. Villalta, W.D. Nes, M. Botta, F.P. Guengerich, G.I. Lepesheva, Sterol 14α-Demethylase Structure-Based Design of VNI ((R)- N-(1-(2,4- Dichlorophenyl)-2-(1 H-imidazol-1-yl)ethyl)-4-(5-phenyl-1,3,4-oxadiazol-2-yl) benzamide)) Derivatives to Target Fungal Infections: Synthesis, Biological Evaluation, and Crystallographic A, J. Med. Chem. 61 (2018) 5679–5691, https:// doi.org/10.1021/acs.jmedchem.8b00641. [142] T.Y. Hargrove, E.P. Garvey, W.J. Hoekstra, C.M. Yates, Z. Wawrzak, G. Rachakonda, F. Villalta, G.I. Lepesheva, Crystal structure of the new in­ vestigational drug candidate VT-1598 in complex with Aspergillus fumigatus sterol 14α-demethylase provides insights into its broad-spectrum Antifungal Activity, Antimicrob. Agents Chemother. 61 (2017), https://doi.org/10.1128/ AAC.00570-17. [143] D.W. Denning, M.J. Bromley, How to bolster the antifungal pipeline, Science (80-. ). 347 (2015) 1414–1416. doi:10.1126/science.aaa6097. [144] M.A. Correia, P.R. Ortiz de Montellano, Inhibition of Cytochrome P450 Enzymes, Cytochrome P450 Struct, Mech. Biochem. (2005) 247–322, https://doi.org/10. 2460/ajvr.72.11.1505. [145] H. Ji, W. Zhang, Y. Zhou, M. Zhang, J. Zhu, Y. Song, J. Lü, J. Zhu, A three-di­ mensional model of lanosterol 14α-demethylase of Candida albicans and its in­ teraction with azole antifungals, J. Med. Chem. 43 (2000) 2493–2505, https://doi. org/10.1021/jm990589g. [146] K. Tasaka, A.; Kitazaki, T.; Tsuchimori, N.; Matsushita, Y.; Hayashi, R.; Okonogi, K. ; Itoh, Optically active antifungal azoles. VII. Synthesis and antifungal activity of stereoisomers of 2-[(1R,2R)-2-(2,4-difluorophenyl)-2-hydroxy-1-methyl-3-(1H-1, 2,4-triazol- 1-yl)propyl]-4-[4-(2,2,3,3-tetrafluoropropoxy)phenyl]-3(2H,4H)- 1,2, 4-triazolone (TAK-, Chem. Pharm. Bull. 45 (1997) 321–326. doi:10.1248/cpb.45. 321. [147] D.M. Rotstein, D.J. Kertesz, K.A.M. Walker, D.C. Swinney, Stereoisomers of Ketoconazole: Preparation and Biological Activity, J. Med. Chem. 35 (1992) 2818–2825, https://doi.org/10.1021/jm00093a015. [148] J. Bartroli, E. Turmo, M. Alguero, E. Boncompte, M.L. Vericat, L. Conte, J. Ramis, M. Merlos, J. Garcia-Rafanell, J. Forn, ChemInform Abstract: New Azole Antifungals. Part 2. Synthesis and Antifungal Activity of Heterocyclecarboxamide Derivatives of 3-Amino-2-aryl-1-azolyl-2-butanol, ChemInform. 29 (2010) no-no, https://doi.org/10.1002/chin.199836174. [149] G. Klopman, D. Ptchelintsev, Antifungal triazole alcohols: A comparative analysis of structure-activity, structure-teratogenicity and structure-therapeutic index re­ lationships using the Multiple Computer-Automated Structure Evaluation (Multi- CASE) methodology, J. Comput. Aided Mol. Des. 7 (1993) 349–362, https://doi. org/10.1007/BF00125508. [150] J. Bartroli, E. Turmo, M. Algueró, E. Boncompte, M.L. Vericat, J. GARCIA- RAFANELL, J. FORN, Synthesis and antifungal activity of new azole derivatives containing an N-acylmorpholine ring, J. Med. Chem. 38 (1995) 3918–3932. doi:10.1021/jm00020a005. [151] D. De Vita, F. Pandolfi, R. Cirilli, L. Scipione, R. Di Santo, L. Friggeri, M. Mori, D. Fiorucci, G. Maccari, R.S.A. Christopher, C. Zamperini, V. Pau, A. De Logu, S. Tortorella, M. Botta, Discovery of in vitro antitubercular agents through in silico ligand-based approaches, Eur. J. Med. Chem. 121 (2016) 169–180, https://doi. org/10.1016/j.ejmech.2016.05.032. [152] L. Friggeri, T.Y. Hargrove, G. Rachakonda, A.D. Williams, Z. Wawrzak, R. Di Santo, D. De Vita, M.R. Waterman, S. Tortorella, F. Villalta, G.I. Lepesheva, Structural basis for rational design of inhibitors targeting Trypanosoma cruzi Sterol 14α-demethylase: Two regions of the enzyme molecule potentiate its in­ hibition, J. Med. Chem. 57 (2014) 6704–6717, https://doi.org/10.1021/ jm500739f. [153] L.M. Podust, T.L. Poulos, M.R. Waterman, Crystal structure of cytochrome P450 14 -sterol demethylase (CYP51) from Mycobacterium tuberculosis in complex with azole inhibitors, Proc. Natl. Acad. Sci. 98 (2002) 3068–3073, https://doi.org/10. 1073/pnas.061562898. [154] F. Chevreuil, A. Landreau, D. Seraphin, G. Larcher, J.P. Bouchara, P. Richomme, Synthesis and antifungal activity of new thienyl and aryl conazoles, J. Enzyme Inhib. Med. Chem. 21 (2006) 293–303, https://doi.org/10.1080/ 14756360600700640. [155] C. Sheng, W. Zhang, H. Ji, M. Zhang, Y. Song, H. Xu, J. Zhu, Z. Miao, Q. Jiang, J. Yao, Y. Zhou, J. Zhu, J. Lü, Structure-based optimization of azole antifungal agents by CoMFA, CoMSIA, and molecular docking, J. Med. Chem. 49 (2006) 2512–2525, https://doi.org/10.1021/jm051211n. [156] J. Bartroli, E. Turmo, M. Algueró, E. Boncompte, M.L. Vericat, L. Conte, J. Ramis, M. Merlos, New Azole Antifungals. 3. Synthesis and Antifungal Activity of 3- Substituted-4(3H)-quinazolinones, J. Med. Chem. 41 (1998) 1869–1882. [157] N.M. Revie, K.R. Iyer, N. Robbins, L.E. Cowen, Antifungal drug resistance: evo­ lution, mechanisms and impact, Curr. Opin. Microbiol. 45 (2018) 70–76, https:// doi.org/10.1016/j.mib.2018.02.005. [158] T.P. Salci, M. Negri, A.K.R. Abadio, T.I.E. Svidzinski, É.S. Kioshima, Targeting Candida spp. to develop antifungal agents, Drug Discov. Today. 23 (2018) 802–814, https://doi.org/10.1016/j.drudis.2018.01.003. [159] A.M. Fuentefria, B. Pippi, D.F. Dalla Lana, K.K. Donato, S.F. de Andrade, Antifungals discovery: an insight into new strategies to combat antifungal re­ sistance, Lett. Appl. Microbiol. 66 (2018) 2–13, https://doi.org/10.1111/lam. 12820. [160] J.A. Parente-Rocha, A.M. Bailão, A.C. Amaral, C.P. Taborda, J.D. Paccez, C.L. Borges, M. Pereira, Antifungal Resistance, Metabolic Routes as Drug Targets, and New Antifungal Agents: An Overview about Endemic Dimorphic Fungi, Mediators Inflamm. 2017 (2017), https://doi.org/10.1155/2017/9870679. M. Shafiei, et al. Bioorganic Chemistry 104 (2020) 104240 21