Download as pdf or txt
Download as pdf or txt
You are on page 1of 18

Expert Opinion on Pharmacotherapy

ISSN: (Print) (Online) Journal homepage: https://www.tandfonline.com/loi/ieop20

An overview of current and emerging antifungal


pharmacotherapy for invasive fungal infections

Scott W. Mueller, Sonya K. Kedzior, Matthew A. Miller, Paul M. Reynolds,


Tyree H. Kiser, Martin Krsak & Kyle C. Molina

To cite this article: Scott W. Mueller, Sonya K. Kedzior, Matthew A. Miller, Paul M. Reynolds,
Tyree H. Kiser, Martin Krsak & Kyle C. Molina (2021): An overview of current and emerging
antifungal pharmacotherapy for invasive fungal infections, Expert Opinion on Pharmacotherapy,
DOI: 10.1080/14656566.2021.1892075

To link to this article: https://doi.org/10.1080/14656566.2021.1892075

Published online: 21 Apr 2021.

Submit your article to this journal

Article views: 98

View related articles

View Crossmark data

Full Terms & Conditions of access and use can be found at


https://www.tandfonline.com/action/journalInformation?journalCode=ieop20
EXPERT OPINION ON PHARMACOTHERAPY
https://doi.org/10.1080/14656566.2021.1892075

REVIEW

An overview of current and emerging antifungal pharmacotherapy for invasive


fungal infections
Scott W. Mueller a,b, Sonya K. Kedziora,b, Matthew A. Millerb, Paul M. Reynoldsa,b, Tyree H. Kisera,b, Martin Krsakc
and Kyle C. Molinab
a
Department of Clinical Pharmacy, University of Colorado Skaggs School of Pharmacy and Pharmaceutical Sciences, Aurora, CO, USA; bDepartment
of Pharmacy, University of Colorado Hospital, Aurora, CO, USA; cDepartment of Medicine, Division of Infectious Diseases, University of Colorado
School of Medicine, Aurora, CO, USA

ABSTRACT ARTICLE HISTORY


Introduction: Invasive fungal infections (IFIs) remain a significant cause of morbidity and mortality Received 16 December 2020
despite significant advancements in currently available therapy. With a flush pipeline of investigational Accepted 15 February 2021
antifungals, the clinician must identify appropriate roles of currently available therapies, potential KEYWORDS
advantages of emerging antifungals, and shortcomings in the evolving clinical evidence. Antifungals; aspergillosis;
Areas covered: Standard and developing treatment approaches for IFIs with currently available anti­ candidiasis; invasive fungal
fungals are summarized with a focus on invasive candidiasis and invasive aspergillosis. Emerging infections; novel antifungal
investigational antifungals are discussed in depth, including mechanisms of action, fungal activity, therapies; pipeline; review
clinical evidence, and ongoing research. An opinion on the impact and potential role of therapy for
emerging antifungals of interest is also provided.
Expert opinion: Despite advances and clinical studies optimizing antifungal use, current therapies
fall short in preventing IFI morbidity and mortality. Further optimization of currently available
antifungals may improve outcomes; however, novel agents are required for historically difficult-to-
treat infections, transitions to oral treatment, minimizing adverse drug effects, decreasing drug
interactions, and ultimately improving patient quality of life. Emerging antifungals may positively
revolutionize the treatment of IFIs.

1. Introduction – invasive fungal infections


triazole resistance exists, the associated mortality rate of
Invasive fungal infections (IFIs) are deep-seated or blood­ IA can exceed 80% [8–10]. Management of IFIs in these
stream-disseminated infections affecting both immunocom­ complex patient populations is challenging with currently
promised and immunocompetent patients [1]. In the available agents due to associated toxicities, drug–drug
intensive care unit (ICU), IFIs make up nearly 20% of micro­ interactions (DDIs), and the development of resistance [11].
biologically documented infections and primarily consist of Three drug classes (triazoles, echinocandins, polyenes)
Candida spp. (~80%), followed by Aspergillus spp. (~10%) [2]. account for the majority of guideline recommended back­
Although IFIs include Cryptococcus spp., dimorphic fungi, bone antifungal therapies for IFIs (Table 1) [12–20].
Mucorales group, Pneumocystis jiroveci, and other hyaline Amphotericin B (AmB), a polyene antifungal, is typically
and dematiaceous molds, the majority of this discussion will used as an intravenous (IV) lipid formulation and demon­
focus on invasive candidiasis (IC) and invasive aspergillosis (IA), strates a wide spectrum of activity, but is associated with
noting niche roles for new therapies in other IFIs. Additionally, significant toxicities including nephrotoxicity, hepatotoxi­
this review of therapeutic approaches will not address current city, and infusion-related reactions. In comparison to AmB,
or novel approaches to diagnostics. For a recent publication triazoles are better tolerated and collectively provide
on diagnostics, we refer the readers to an excellent publica­ a broad spectrum of activity with oral formulations avail­
tion by the Mycoses Study Group and European Organization able. However, triazoles are associated with significant
for Research and Treatment of Cancer [3]. DDIs and hepatotoxicity. Echinocandins offer fungicidal
Associated mortality rates for IC (specifically candide­ activity against yeast and are well tolerated but are nar­
mia) are approximately 25%, but can reach as high as row-spectrum and lack oral formulations. We refer the
40–60% in the ICU population [4–6]. Although IA is often reader to an in depth review of current antifungal activity,
associated with neutropenic and hematologic malignan­ labeled and off-labeled use, and clinical pearls published
cies, it has been increasingly recognized as a pathogen in elsewhere [19]. To reduce the high mortality rates seen
non-neutropenic hosts [7]. In some populations, where with IFIs, novel antifungals which circumvent these

CONTACT Scott W. Mueller scott.mueller@cuanschutz.edu Department of Clinical Pharmacy, University of Colorado Skaggs School of Pharmacy and
Pharmaceutical Sciences, Aurora, CO, USADepartment of Pharmacy, University of Colorado Hospital, Aurora, CO, USA
*
Co-first/lead author SWM and SKK
© 2021 Informa UK Limited, trading as Taylor & Francis Group
2 S. W. MUELLER ET AL.

Candida spp. have become increasingly resistant with


Article highlights approximately 7% of bloodstream isolates demonstrating flu­
conazole resistance, the majority being Candida glabrata and
● There is an unmet clinical need to address emerging antifungal Candida krusei [6]. Voriconazole is an acceptable alternative for
resistance among invasive fungal infections (IFIs).
● Significant advancements in the antifungal pipeline have resulted in voriconazole-susceptible isolates. However, its non-linear
promising agents with unique mechanisms of action, improved for­ pharmacokinetic (PK) profile is characterized by extensive
mulations, fewer toxicities, and drug–drug interactions compared to interindividual variability subject to CYP-2C19 polymorphisms
many current antifungal therapies.
● Tetrazoles, specifically oral oteseconazole, VT-1129, and VT-1598, and coupled with significant toxicities (e.g., CNS, hepatotoxi­
have increased specificity for fungal CYP51 and the potential to city, periostitis, squamous cell carcinoma) can limit use [24].
replace triazoles in many fungal infections (e.g., vulvovaginal candi­ Echinocandin resistance has also been rising within
diasis) as they are associated with minimal drug–drug interactions
and fewer adverse effects. Tetrazoles can preserve excellent activity C. glabrata, often leaving only toxic and poorly tolerated
against some resistant IFIs and boast a broad spectrum of activity, alternatives [6,12,23].
particularly with VT-1598. The greatest concern among Candida spp. is the increasing
● Rezafungin is a new echinocandin with an extended half-life allowing
for once weekly intravenous dosing while maintaining a satisfactory rate of resistant non-albicans infections [25,26]. Candida auris
pharmacokinetic/pharmacodynamic and safety profile against is rapidly emerging globally as a multidrug-resistant (MDR)
Candida, Aspergillus and Pneumocystis spp. yeast that has demonstrated an increase in United States
● Ibrexafungerp is a novel oral and intravenous triterpenoid with
a broad spectrum and has shown promising efficacy in triazole and (U.S.) cases by over 300% in 2018 compared to 2015–2017
echinocandin-resistant infections. In vivo and in vitro activity has been [22]. The Centers for Diseases Control and Prevention reported
demonstrated for the treatment of invasive candidiasis, invasive that 90% of the isolates were resistant to at least one anti­
aspergillosis and other fungal infections refractory to current stan­
dards of care. fungal, 30% were resistant to at least two, and approximately
● Encochleated amphotericin B, pending clinical efficacy studies, is en 4% of the isolates have been resistant to the three aforemen­
route to be the first oral amphotericin maintaining a broad range of tioned drug classes [22,27]. Echinocandins remain a preferred
activity against yeasts and molds with reduced toxicity compared to
intravenous formulations. empiric therapy with antifungal combinations being required
● Fosmanogepix is formulated for both oral and intravenous adminis­ in cases of MDR, unresponsive infections, or difficult-to-treat
tration against multidrug-resistant and difficult-to-treat infections sites [12,28]. Additional therapies for this increasingly resistant
including Candida spp., Aspergillus spp., and rare molds. It has
a favorable safety profile and attains wide tissue distribution, making pathogen with associated mortality rates of 30–78% in IC are
it a viable future option for empiric treatment and continuation as urgently needed [22,29].
oral step-down therapy.
● Olorofim offers a unique mechanism by inhibiting fungal growth via
inhibition of dihydroorotate dehydrogenase. It has shown efficacy in
targeting Aspergillus spp. and rare molds, but is limited in its yeast
coverage. This agent has great promise as a targeted mold agent or 2.2. Invasive Aspergillosis (IA)
for salvage therapy in refractory or difficult-to-treat IFIs.
Patient outcomes following IA remain poor despite clear
This box summarizes key points contained in the article. expert guidance for antifungal therapies targeting Aspergillus
spp. The IDSA Aspergillosis Guidelines recommend voricona­
zole for IA as a preferred therapy, with liposomal AmB or
isavuconazole as alternatives [14,30]. European guidelines
recommend either voriconazole or isavuconazole as initial
limitations are urgently needed. This review provides an treatment for IA, but advocate for switching to liposomal
update on antifungal treatment, focusing on new and AmB if voriconazole resistance is suspected or confirmed in
emerging agents and their place in therapy for IFIs. Aspergillus fumigatus or in those unresponsive to initial mold-
active triazoles (Figure 2) [15].
Combinations of echinocandins and triazoles have
2. Overview of current antifungal approaches demonstrated synergy in vitro against many Aspergillus
spp. For difficult-to-treat IA, voriconazole in combination
2.1. Invasive Candidiasis (IC)
with an echinocandin could be considered [14,15].
In selected hospitalized populations, Candida spp. remains the However, clinical trials with combination therapies have
fourth most common cause of bloodstream infections [12 not shown consistent clinical superiority over appropriate
,21–23]. Current therapeutic strategies have been reviewed triazole monotherapy [28,31]. In patients with hematologic
in depth within the Infectious Disease Society of American malignancies with IA, 6-week mortality was non-
(IDSA) guidelines [12]. Echinocandins are recommended for significantly lower with voriconazole–anidulafungin combi­
initial treatment in critically ill and neutropenic patients with nation compared to voriconazole alone (19.3% vs. 27.5%,
candidemia, and are effective for various IC infections with the p= 0.087) [31]. Combination therapy may be useful for
exception of central nervous system (CNS) and urinary infec­ patients with high fungal burden, suboptimal response to
tions. Lipid formulations of AmB are an option for echinocan­ monotherapy, or as salvage therapy, since high failure
din-resistant or refractory IC, or for deep-seated infection sites rates have been reported in the treatment of IA with
where echinocandins lack adequate penetration. Stepdown echinocandin monotherapy [28].
therapy to a triazole is possible once candidemia has cleared, Azole-resistance has also been increasing worldwide
susceptibilities are confirmed, and clinical improvement is [26,32]. Azole-resistant A. fumigatus (ARAF) threatens the
observed (Figure 1). effectiveness of mold-active triazoles, which are currently
Table 1. Selected currently available anti-fungal drug classes [12,14,15].
Strategies for
Class Target Agents TDM (mg/L) Advantages Current limitations improvement
Triazoles Inhibit 14α-lanosterol ● Fluconazole Trough Levels ● Oral formulations ● DDIs ● Improved fungal
demethylase ● Itraconazole ● Well tolerated compared to polyenes ● Toxicities (hallucinations, hepatotoxicity, enzyme selectivity
● Voriconazole:
● Voriconazole ● Agent specific extended spectrum QT prolongationb) (CYP51)
● Posaconazole 1–5.5 ● Favorable pharmacokinetics, tissue, and ● Erratic absorption of some oral ● Optimize formulations
● Itraconazole:
● Isavuconazole CNS distribution (for many triazoles) formulations to improve absorption
1–4 ● Non-linear PK (TDM recommended for
● Posaconazole: itraconazole, voriconazole, posaconazole)
1–3.75
● Isavuconazole:
2–3a
Glucan Inhibit 1,3-βD-glucan ● Anidulafungin N/A ● Excellent activity against Candida spp. ● IV formulations only ● Oral formulation
Synthesis synthesis ● Caspofungin ● Fungicidal ● Once daily dosing ● Longer t1/2
Inhibitors ● Micafungin ● Wide spectrum ● Minimal distribution to CNS, urine ● Improved CNS distribu­
● Optimal safety profile tion
● Minimal DDIs
Polyenes Inhibit ergosterol binding ● Amphotericin B (deoxycholate, N/A ● Broad spectrum ● Toxicities (infusion reactions, ● Decreased systemic
to fungal membrane lipid, and liposomal formulations) ● Fungicidal nephrotoxicity, hepatotoxicity) toxicities
● CNS tissue distribution for most formu­ ● IV formulation only ● Oral formulation
lations

CNS: central nervous system; DDI: drug–drug interactions; IV: intravenous; N/A: not applicable; PK: pharmacokinetic; TDM: therapeutic drug monitoring; t1/2: half-life
a
Provisional target, TDM not universally recommended and may be unavailable
b
Isavuconazole is QT shortening
EXPERT OPINION ON PHARMACOTHERAPY
3
4 S. W. MUELLER ET AL.

Figure 1. Working framework of antifungal therapy for disseminated candidiasis with currently available agents as empiric therapy and possible roles for future
agents.
a) Clinically stable non-neutropenic patients. Preferred for C. parapsilosis, C. krusei is fluconazole resistant, and C. glabrata may have higher MICs or resistance to triazoles.
b) Echinocandins preferred empiric therapy for most including clinically unstable and/or neutropenic patients. C. parapsilosis has higher echinocandin MICs.
c) Lipid or liposomal AmB formulation is an alternative agent to those with first-line intolerance or suspected or proven resistance to other agents but is limited by toxicities. Liposomal
products are preferred in some scenarios. C. lusitaniae may be AmB resistant.
d) Clinical response good: Partial to complete resolution of clinical symptoms, clearance of cultures, decrease in fungal biomarkers (e.g., 1, 3 Beta-D-glucan) and improvement in radiological
data (>25–50% decrease in lesion size if available).
e) Clinical response poor: Minimal to no improvement on current therapy based on clinical, radiological, and microbiological data.
f) Clinically appropriate: Patient is clinically stable and tolerating oral.
g) Potential causes of poor response: Source control, PK/PD, inadequate dose/penetration to site of infection, failure to meet TDM targets, etc.
h) Often retains activity against triazole-resistant organisms, however, elevated MICs may occur.
i) APX001: No activity against C. krusei, higher MIC C. kefyr.
j) CAmB if AmB susceptible. AmB: Amphotericin-b; APX001: Fosmanogepix; CAmB: Encochleated amphotericin B; IBX: Ibrexafungerp; L-AmB: Lipid or liposomal Amphotericin-b product; PD:
Pharmacodynamic; PK: Pharmacokinetic; RZA: Rezafungin.

the preferred initial and only oral therapeutic options. The overcome elevations in minimum inhibitory concentrations
Netherlands has documented increasing prevalence in ARAF (MICs). Such approaches require close TDM and subse­
isolates from 2013 to 2018 (7.6% to 14.7%, p= 0.0001), and quent dose adjustments, which may be burdensome or
although reports of ARAF are increasing in the U.S., they not available in all settings [39].
remain relatively uncommon [22,33,34]. Importantly, mortal­ Posaconazole retains the most in vitro activity against
ity rates increase from 40% with susceptible strains to ARAF, and in vivo studies suggest that it may achieve PD
a staggering 90% with resistant strains [35]. Triazole resis­ targets in isolates with MICs of 0.5 mg/L (European
tance epidemiology, mechanisms, and detection in Committee on Antimicrobial Susceptibility Testing
Aspergillus spp. have been reviewed in detail elsewhere [EUCAST] breakpoint of 0.125 mg/L) [40,41]. There is inter­
[1,14,36]. est in augmenting posaconazole exposure to overcome
elevated MICs, which is supported by PK modeling
[23,41,42]. Ninety-percent target attainment (AUC0-24/MIC
2.3. Optimizing current triazoles ≥167) was obtained with standard IV and delayed-release
The increasing rates of ARAF and limited second-line posaconazole dosing at an MIC of 0.125 mg/L, but
options for IA emphasize the importance of triazole use a cumulative fractional response of <80% was predicted
optimization. This goal is complicated by extensive intra- for all simulated Aspergillus spp., except for Aspergillus
and inter-patient PK and pharmacodynamic (PD) variability; terreus. High-dose steady state regimens (200 mg twice
therefore, therapeutic drug monitoring (TDM) to meet tar­ daily) may be needed to meet cumulative fractional
gets safely is paramount (Table 1) [37]. In clinical practice, response PD efficacy targets for Aspergillus flavus,
triazole trough concentrations are used as surrogate mar­ Aspergillus fumigatus, and Aspergillus nidulans.
kers for area under the curve (AUC), the PD parameter Additionally, a clinical case series suggests that troughs
associated with improvements in clinical effectiveness >3 mg/L may be a reasonable target to overcome ARAF
[38]. In situations of variable triazole susceptibility, choos­ infections [42]. More clinical data are needed, but TDM-
ing an azole that can meet PK/PD targets through opti­ guided higher posaconazole exposures as a step-down
mized dose-escalation, without undue toxicity, may help therapy (with or without an echinocandin) could improve
EXPERT OPINION ON PHARMACOTHERAPY 5

Figure 2. Working framework of antifungal therapy for invasive aspergillosis with currently available agents as empiric therapy and possible roles for future agents.
a) Triazoles (voriconazole or isavuconazole) are preferred first-line therapy for most invasive aspergillus infections.
b) Combination triazole-echinocandin preferred empiric therapy if regional A. fumigatus triazole resistance >10% (some experts recommend if >5%). Synergistic combination trended
toward improved outcomes in some populations. Two weeks of combination therapy trended toward improved outcomes vs. triazole monotherapy among those with hematologic
malignancy and invasive aspergillosis, though some situations may require longer combination therapy (e.g., resistance).
c) Liposomal Amphotericin-b an alternative preferred empiric therapy if regional A. fumigatus triazole resistance >10% but is limited by toxicities. Not effective for A. terreus.
d) Clinical response good: Partial to complete resolution of clinical symptoms, clearance of cultures, decrease in fungal biomarkers (e.g., galactomanin) and improvement in radiological
data (>25–50% decrease in lesion size if available).
e) Clinical response poor: Minimal to no improvement on current therapy based on clinical, fungal biomarkers, radiological, and microbiological data.
f) ISA TDM controversial, target trough not set.
g) Clinically appropriate: Patient is clinically stable and tolerating oral.
h) Potential causes of poor response: Source control, PK/PD, inadequate dose/penetration to site of infection, failure to meet TDM targets, and so on.
i) VT-1598 as the tetrazole active against Aspergillus spp. Elevated VT-1598 MIC and resistance are reported for triazole resistant A. fumigatus.
j) CAmB if AmB susceptible. AmB: Amphotericin-b; CAmB: Encochleated amphotericin B; APX001: Fosmanogepix; IBX: Ibrexafungerp; ISA: Isavuconazole; LAmB: Liposomal amphotericin-b;
OLO: Olorofim; PD: Pharmacodynamic; PK: Pharmacokinetic; POS: Posaconazole; RZA: Rezafungin; TDM: Therapeutic drug monitoring.

outcomes in patients with ARAF with posaconazole MICs of sparing mammalian CYP450 enzymes [45]. Such selectivity
0.5 mg/L; however, aggressive dosing may incur additional could minimize CYP450-mediated DDIs and hepatotoxicity,
toxicities [39,43]. Similar PK simulations suggest that dou­ which limits current triazole use. Three tetrazoles under inves­
bling the standard dose of isavuconazole could achieve tigation will be discussed: oteseconazole, VT-1129, and VT-
90% target attainment for ARAF with MICs of 2 mg/L, 1598.
although more data are needed confirming safety at
higher dosages [44].
3.1.1. Oteseconazole
Oteseconazole (previously VT-1161), is an oral tetrazole anti­
3. Emerging antifungals fungal currently in Phase 2 and Phase 3 clinical development
for the treatment of onychomycosis and recurrent vulvova­
Fortunately, promising novel antifungal agents and classes to ginal candidiasis (RVVC), respectively [46–49]. Oteseconazole
overcome drug resistance achieve adequate concentrations has potent in vitro activity against Candida spp. with geo­
at the site of infection and provide oral options for prolonged metric mean MICs lower than that of compared triazoles [50].
treatment courses while minimizing toxicity and potential It retains activity in many fluconazole-resistant C. glabrata
DDIs, are in the pipeline. Ideal agents have high selectivity strains; however, experimental MICs increase with ABC-
for fungal cells, minimal off-target effects, and potent activity transporter overexpression, suggesting that azole cross resis­
against IFIs to decrease morbidity and mortality (Tables 2 and tance is possible [51].
3; Figures 1 and 2) [1]. Compared to fluconazole, oteseconazole showed non-
statistical improved efficacy for the treatment of VVC with
low mycological recurrence rates at 3 and 6 months [52].
3.1. Tetrazoles The median reported half-life was on the order of months.
Tetrazoles, like the triazoles, inhibit fungal cell wall synthesis A Phase 2b placebo-controlled dose-ranging study sup­
by preventing the conversion of lanosterol to ergosterol cata­ ported low rates of 48-week recurrences when given weekly
lyzed by 14α-demethylase. In contrast, tetrazoles bind fungal for 11 weeks [53]. Active Phase 3 studies will provide more
CYP51 with much higher selectivity (>2,200-fold), largely definitive clinical data regarding oteseconazole 150 mg
6 S. W. MUELLER ET AL.

weekly for RVVC [47–49]. Oteseconazole has been granted probability of DDIs, position tetrazoles as a promising new
Qualified Infectious Disease Product and fast track designa­ antifungal class.
tions by the Food and Drug Administration (FDA) and may
be the first tetrazole to receive FDA-approval (Table 4) [1].
3.2. Glucan synthase inhibitors
3.1.2. VT-1129 Echinocandins are glucan synthase inhibitors which prevent
VT-1129 showed in vitro activity against 31 Candida spp., the biosynthesis of β-1,3-glucan, a key component of the
including azole-resistant strains, in patients with chronic fungal cell wall structure [60,61]. There are currently three
mucocutaneous candidiasis [54]. An in vitro study tested VT- FDA approved echinocandins for the treatment of candidemia
1129 and oteseconazole activity against azole and echinocan­ and IC: caspofungin, micafungin, and anidulafungin. Two glu­
din-resistant C. krusei (n = 50) and C. glabrata (n = 34) isolates can synthesis inhibitors are under investigation that offer
[55]. The MIC50 for VT-1129 and oteseconazole against C. krusei improved PK and PD profiles; rezafungin, a novel once-
were 0.5 and 0.25 mg/L, respectively, and 0.12 mg/L for both weekly echinocandin, and ibrexafungerp a triterpenoid, glucan
agents against C. glabrata. Although clinical breakpoints are synthase inhibitor with a differentiated structure and enzyme-
not defined, all isolates were inhibited at concentrations inhibition properties.
≤2 mg/L of VT-1129 and oteseconazole, again demonstrating
potency even in the setting of resistance. 3.2.1. Rezafungin
VT-1129 may be especially useful against Cryptococcus Rezafungin (CD101, previously SP3025), is an IV echinocandin
spp. It retained great activity against investigator labeled under Phase 3 investigation for the treatment of IC including
“fluconazole dose-dependent susceptible” (n = 27) and candidemia and prophylaxis of invasive fungal disease in
“resistant” (n = 6) Cryptococcus neoformans isolates with patients undergoing allogeneic blood and marrow transplan­
MIC50 ranges of 0.05 and 0.25 mg/L, respectively [56]. This tation [62,63]. Rezafungin has received Qualified Infectious
is in comparison to fluconazole’s dose-dependent MICs Disease Product and fast track status by the FDA for these
16–32 mg/L, and resistant MICs ≥ 64 mg/L. A murine model indications [1].
demonstrated successful fungal burden reduction and survi­ Rezafungin has enhanced stability, solubility, and pene­
val with VT-1129 compared to fluconazole and placebo trates difficult-to-treat sites, such as infected abscesses, but
against C. neoformans [57]. VT-1129 showed 100% survival like other echinocandins, it has limited CNS and urine
with undetectable fungal burden more than 20 days after penetration [64,65]. Promisingly, rezafungin has
regimen completion, whereas rebound fungal burden was a prolonged half-life (~133 hours) allowing for once-
observed in the fluconazole arm. weekly dosing [64,66]. This offers an improved PD profile
through front loading, where rapid fungal killing is
3.1.3. VT-1598 achieved with high initial concentrations, especially in
VT-1598 is an oral agent with the broadest antifungal activ­ hard-to-penetrate infectious sites, and may decrease the
ity among the three investigational tetrazoles. It is active potential for resistance development.
against yeasts, molds, endemic fungi, and retains in vitro Rezafungin provides potent in vitro activity against
activity against many resistant isolates, such as a variety of Candida spp. including azole-resistant and MDR strains
Candida spp. (n = 175) and Aspergillus spp. (n = 134) [58]. [67]. The in vitro activity of rezafungin was evaluated in
The MIC50 for all Candida spp. were lower than that of the SENTRY surveillance program, which included 1,904
fluconazole. Specifically for C. glabrata, the MIC50/100 was Candida isolates collected worldwide between 2016 and
0.248/6.04 mg/L and it retained activity against many azole- 2018 [68]. Rezafungin had comparable activity to anidula­
resistant strains, MIC50/100 1.19/>8 mg/L. VT-1589 has also fungin against Candida albicans (n = 835; MIC90 0.06 vs.
shown compelling in vitro activity for Aspergillus spp. with 0.03 mg/L), C. glabrata (n = 374; MIC90 0.12 vs. 0.12 mg/L),
geometric mean MIC ranges similar to posaconazole and Candida tropicalis (n = 196; MIC90 0.06 vs. 0.06 mg/L) and
voriconazole for A. flavus (0.685 mg/L), Aspergillus niger C. krusei (n = 77; MIC90 0.06 vs. 0.125 mg/L). Rezafungin
(1.78 mg/L), and A. terreus (0.533 mg/L). In wild-type isolates had greater in vitro activity against C. auris isolates (n = 16)
of A. fumigatus, geometric mean MIC ranges of VT-1598 compared to caspofungin and micafungin, but equivocal
(0.25–2 mg/L) tended to be similar to that of posaconazole potency to anidulafungin [25,69]. Rezafungin demonstrated
and voriconazole. However, in ARAF isolates containing similar activity to anidulafungin against A. fumigatus
CYP51A mutations, the MIC of VT-1598 significantly (n = 183; MIC90 0.03 mg/L) and A. flavus (n = 45; MIC90
increased (MIC100 = 13.3 mg/L) suggesting cross-resistance. 0.015 mg/L) [68].
For C. auris (n = 100), VT-1598 had MIC ranges between A Phase 2, randomized, double-blinded, multicenter
0.03 and 8 mg/L (mode 0.25 mg/L) with activity in fluconazole- study (STRIVE), compared IV rezafungin 400 mg once weekly
resistant strains and improvements in fungal burden similar to (400 mg; n = 81), rezafungin 400 mg on week 1 followed by
caspofungin [59]. Future clinical and PK/PD studies will help 200 mg weekly (400/200 mg; n = 57), and caspofungin
define the role of VT-1598 in the treatment of C. auris; how­ 70 mg once, followed by 50 mg daily (70/50 mg; n = 69)
ever, current data suggests it retains activity in isolates with for the treatment of candidemia and IC [70]. The overall
reduced susceptibilities to currently available regimens. cure rate at day 14 was non-statistically higher in the reza­
Overall, the potency and activity against a broad array of fungin 400/200 mg weekly regimen (76.1%) compared to
fungal species, including resistant organisms, and decreased 400 mg weekly (60.5%) and caspofungin (67.2%). All-cause
EXPERT OPINION ON PHARMACOTHERAPY 7

mortality at day 30 was lowest in the 400/200 mg group MICs for ibrexafungerp (0.06 to 2 mg/L) were comparatively
(4.4%) versus 400 mg (15.8%) and caspofungin (13.1%). The narrower than ranges for anidulafungin (0.03 to >32 mg/L)
safety analysis showed comparable rates of adverse events and micafungin (0.016 to >32 mg/L). Of the eight echinocan­
across all arms. din-resistant isolates with FKS mutations, ibrexafungerp had
Rezafungin is currently under investigation in multiple MICs of 0.25 mg/L (n = 3) or 0.5 mg/L (n = 5). Currently
Phase 3 trials. The ReSTORE trial is a non-inferiority study assessed FKS mutations have not affected ibrexafungerp to
comparing rezafungin 400/200 mg weekly to caspofungin the same degree as echinocandins, likely due to nonidentical
70/50 mg daily for the treatment of IC including candidemia binding sites of action [78,79]. Similar MIC ranges have been
[62]. The ReSPECT trial is investigating rezafungin 400/ seen in other in vitro studies in C. auris isolates [72,78,80,81].
200 mg weekly in prevention of Candida, Aspergillus and A Phase 2 study evaluated the safety and efficacy of ibrex­
Pneumocystis infections in patients undergoing allogeneic afungerp therapy in non-neutropenic patients (n = 21) with IC
blood and marrow transplant (Table 4) [63]. Further clinical [82]. After an initial echinocandin treatment for 3–10 days,
data are needed to determine the role of rezafungin for patients were randomized either to oral ibrexafungerp
Pneumocystis prophylaxis or treatment, but murine data 1000 mg then 500 mg daily (1000/500 mg; n = 6) or ibrex­
appear promising [71]. afungerp 1250 mg then 750 mg daily (1250/750 mg; n = 7)
and were compared to standard of care (SOC) (n = 8). Similar
3.2.2. Ibrexafungerp rates of global favorable responses were seen across all treat­
Ibrexafungerp (SCY-078, previously MK-3118) is a novel, triter­ ment arms, 1000/500 (71%), 1250/750 (86%) and SOC (71%).
penoid, glucan synthase inhibitor currently undergoing late- No differences in adverse events were noted between the
phase clinical trials (Table 4) [67]. It is being developed as both groups. Population PK analysis demonstrated that the 1250/
oral and IV formulations, allowing for flexibility in dosing regi­ 750 mg dosing regimen achieved the greatest steady-state
mens. In vitro and in vivo data have demonstrated potent target exposure, supporting this dose for further clinical
activity against MDR Candida and Aspergillus spp., and is cur­ evaluation.
rently being evaluated for IC and IA [72–74]. SCYNERGIA is a Phase 2 clinical trial evaluating ibrexafun­
Ibrexafungerp is a substrate of CYP3A4 and a potential gerp in combination with voriconazole versus voriconazole
inhibitor of CYP2C8 in vitro, but no significant DDIs have monotherapy for the treatment of IA [83]. Ibrexafungerp dos­
been identified to preclude clinical use [75]. Unlike echinocan­ ing is 500 mg orally twice daily for 2 days, followed by 500 mg
dins, ibrexafungerp achieves tissue concentrations several-fold daily for a minimum of 6 weeks. Preclinical animal models
higher than plasma concentrations, which may be beneficial have shown lower pulmonary infarct scores and improved
for IFIs where high tissue penetration is required, such as in survival. This combination was proven synergistic against
the lung, liver, bone, or vaginal tissue [76]. Similar to echino­ Aspergillus isolates (n = 51) from lung transplant patients [84].
candins, ibrexafungerp is unlikely to adequately penetrate the FURI is a Phase 3 single-arm, open-label trial of oral ibrex­
CNS or urine, but is expected to have minimal off-target afungerp in those with refractory IFIs or intolerant to SOC [85].
effects limiting potential toxicities. The hospital-based regimen is 750 mg orally twice daily for 2
The in vitro activity of ibrexafungerp against wild-type, days, followed by 750 mg daily for up to 6 months. At an
azole-resistant, and echinocandin-resistant Candida isolates interim analysis, 83% (n = 34) of patients have experienced
compared to caspofungin and fluconazole revealed the fol­ a clinical benefit of either a complete or partial (56%; n = 23)
lowing respective MIC90 values: C. albicans (n = 29; 1, 2, or stable (27%; n = 11) response [84]. The most common
≥128 mg/L), C. glabrata (n = 29; 2, 16, ≥128 mg/L), Candida adverse events were mild to moderate gastrointestinal (GI)
parapsilosis (n = 15; 0.5, 0.5, 64 mg/L), C. tropicalis (n = 21; 1, 1, disturbances, which were generally tolerated.
≥128 mg/L), and C. krusei (n = 19; 2, 1, ≥128 mg/L) [73]. CANDLE is a Phase 3 multicenter, randomized, double-
Ibrexafungerp MIC90 was 8-fold lower than caspofungin for blinded study evaluating the efficacy and safety of oral ibrex­
C. glabrata strains, and remained highly potent in fluconazole- afungerp in women with RVVC compared to placebo [86].
resistant strains. In vitro activity of ibrexafungerp was also Patients with response to fluconazole for acute treatment of
compared against AmB and caspofungin for Aspergillus spp. VVC will be randomized to receive ibrexafungerp 300 mg
revealing the following respective MIC90 values: A. flavus orally twice daily for 1 day or placebo. If they fail fluconazole
(n = 23; 0.12, 2, 0.03 mg/L), A. fumigatus (n = 21; 0.25, 2, treatment, they will be enrolled in an open-label sub-study
0.06 mg/L), A. terreus (n = 18; 0.12, 2, 0.06 mg/L), and MIC50 and receive ibrexafungerp 300 mg twice daily until their acute
A. niger (n = 9; 0.06, 1, 0.03 mg/L) [74]. Ibrexafungerp’s episode has been resolved. The primary endpoint is RVVC at
potency was not superior to caspofungin, but displayed ade­ 24 weeks of evaluation.
quate activity to make it a viable option pending further CARES is a Phase 3 single-arm, open-label, emergency pro­
clinical research. Preliminary studies also suggest that ibrexa­ tocol study assessing ibrexafungerp in those with IC or candi­
fungerp could be utilized in Pneumocystis based on a murine demia due to C. auris [87]. Ibrexafungerp is dosed at 750 mg
and prophylaxis model [1]. orally twice daily for 2 days, then 750 mg once daily for up to
Ibrexafungerp has in vitro activity against C. auris strains 90 days. Complete response has been reported in two patients
(n = 122), including echinocandin-resistant isolates [77]. The with candidemia following 17 and 22 days of therapy [88].
8 S. W. MUELLER ET AL.

3.3. Polyenes Although there is a high cost associated with manufactur­


ing cochleates and other potential limitations, the prospect of
AmB is an IV polyene antifungal that disrupts the integrity of the
safely delivering a highly active antifungal otherwise known
fungal cell membrane by interacting with ergosterol, causing
for toxicities orally to the site of active infection remains
cellular membrane leakage and releasing intracellular compo­
attractive [90].
nents leading to cell death [60,61]. AmB retains broad fungicidal
activity, but tolerability remains poor despite various IV lipid
formulations. To circumvent these limitations, a novel nanopar­
ticle-based encochleated AmB has been developed as an orally 3.4. Glycosylphosphatidylinositol (GPI) inhibitors
bioavailable polyene formulation [89]. In the novel class of GPI inhibitors, fosmanogepix (APX001) is
a prodrug of manogepix (E121) that inhibits the fungal
3.3.1. MAT2203 enzyme Gwt1 [97,98]. This leads to inactivation of GPI anchor
Encochleated AmB (CAmB; MAT2203) utilizes a nanostructure proteins thereby decreasing the integrity of the fungal cell
chelate allowing for water-insoluble or hydrophobic drugs to wall. This differentiated mechanism of action is specific to
be carried and delivered from the GI tract to the systemic fungal cells thereby limiting adverse effects to human cells.
circulation. There it can undergo macrophagic endocytosis,
releasing the active drug to the fungal cell wall using macro­ 3.4.1. Fosmanogepix
phages for transportation [1,90]. CAmB has been granted FDA Fosmanogepix is a broad-spectrum antifungal with activity
fast track, Qualified Infectious Disease Product, and orphan against yeasts, such as Candida spp. (except C. krusei, poten­
drug designations for the treatment of IC and IA, prevention tially Candida kefyr, but including C. auris), C. neoformans, and
of IFIs in immunocompromised patients, and treatment of Coccidioides immitis. Additionally, its activity extends to molds
cryptococcosis [1]. such as Aspergillus, Fusarium, and Scedosporium spp. with less
In vivo models suggest CAmB rapidly attains high con­ potency, but in vivo activity against some of the Mucorales
centrations in target organs such as the liver, spleen, and group [97,99]. Fosmanogepix is currently in three Phase 2 trials
kidneys [91,92]. Mice infected with C. albicans were treated for IC, C. auris, IA, and is in pre-clinical status for
with oral CAmB 0.5–10 mg/kg/day or intraperitoneal AmB Cryptococcosis [1, 67].
1–2 mg/kg/day [92]. While all CAmB and AmB 2 mg/kg Phase 1 clinical trials have shown excellent safety and
dosing strategies resulted in 100% survival, tissue concen­ tolerability profiles along with superior bioavailability (>90%),
trations exceeded the MIC of 0.25 mg/L in the 10 mg/kg allowing for convenient IV to oral transitions [1,97].
CAmB group after 1–2 days versus 3–5 days for AmB. Tissue Fosmanogepix has also shown adequate tissue penetration
concentrations with CAmB were maintained slightly above into the brain, lung, kidney and eye, while clinically relevant
the MIC, whereas AmB increased by 4 to 40-fold by day 15. DDIs have remained low [100].
This suggests that CAmB can rapidly attain high concentra­ In vitro studies have shown equivocal to superior activity
tions in targeted tissues, without accumulation, thereby of fosmanogepix compared to echinocandins and triazoles
mitigating potential side effects or toxicities. In a murine [101]. The MIC90 values for fosmanogepix, anidulafungin and
model of disseminated aspergillosis, CAmB at 40 mg/kg voriconazole against Candida spp. are as follows, respec­
attained 70% survival versus 20% in the intraperitoneal tively: C. albicans (n = 414; 0.008, 0.03, 0.015 mg/L),
AmB 4 mg/kg group [93]. Further, dose ranging studies C. glabrata (n = 321; 0.12, 0.12, 1 mg/L), and C. parapsilosis
are warranted, but in vivo data are promising. (n = 270; 0.015, 2, 0.06 mg/L). Fosmanogepix was less active
Phase 1 tolerability studies suggest that daily divided doses against C. krusei (n = 43; >2, 0.12, 0.5 mg/L) and C. kefyr
of 1–2 g/day are tolerable, and doses of 1.5 g/day for a week (n = 12; 0.5, 0.12, ≤0.008 mg/L). Fosmanogepix activity
showed no renal toxicity in 9 patients [94]. A Phase 2 study of against Aspergillus spp. was comparable to anidulafungin
patients with moderate to severe VVC evaluated the safety and more potent than voriconazole with these respective
and efficacy of CAmB (200 mg; n = 46 and 400 mg; n = 45) for MIC90 values: A. fumigatus (n = 182; 0.03, 0.03, 0.5 mg/L),
5 days compared to a single dose of fluconazole (150 mg; A. flavus (n = 18; 0.03, 0.03, 1 mg/L), A. niger (n = 23; 0.015,
n = 46) [95]. Clinical cure, mycological response, and overall 0.015, 2 mg/L), and A. terreus (n = 10; 0.03, 0.03, 0.5 mg/L).
response rates were inferior with CAmB. Although GI side For C. auris isolates (n = 16), fosmanogepix MIC90 values
effects were higher with CAmB (22–27%) compared to fluco­ were 8-fold lower than anidulafungin (0.031 vs. 0.25 mg/L) and
nazole (9%), the lack of nephrotoxicity and hepatotoxicity after >30-fold lower than voriconazole (0.031 vs. 1 mg/L) [102]. In
a short course of therapy was importantly recognized. vivo data in a neutropenic mouse model (n = 5) showed that
EnACT is a Phase 2 prospective study investigating the intraperitoneal fosmanogepix 78 mg/kg three times daily had
safety, efficacy and tolerability of CAmB for the treatment of significantly higher survival outcomes than intraperitoneal
cryptococcal meningitis in approximately 140 HIV-infected anidulafungin 10 mg/kg twice daily (100% vs. 50%;
patients (Table 4) [96]. The study will be rolled out in four p= 0.034). Fosmanogepix maintained better CNS penetration
phases and is planned to compare CAmB to SOC as stepdown based on a 1.48 log reduction in colony-forming unit counts
and initial therapy [1,96]. The primary efficacy endpoint at day after 48 hours of treatment (p= 0.005).
14 will assess the reduction in fungal cell counts measured by In a multicenter, open-label Phase 2 trial (SURGE), the safety
cerebral spinal fluid studies, which is pivotal in providing CNS and efficacy of fosmanogepix were evaluated for first-line
activity data. treatment of candidemia in non-neutropenic patients
EXPERT OPINION ON PHARMACOTHERAPY 9

(n = 21) with suspected or confirmed antifungal-resistance a midazolam probe study concluded a weak inhibitory effect
[103]. Patients were excluded if they were colonized with on CYP3A4 [107].
C. krusei or had deep-seated candidal infections. Patients Olorofim has excellent in vitro potency against azole-
were treated for up to 14 days with fosmanogepix 1000 mg resistant and difficult-to-treat Aspergillus isolates (n = 213).
IV twice daily on day one, followed by 600 mg IV once daily, Respective MIC90 values of olorofim, voriconazole, and anidu­
which could transition to 700 mg orally once daily after day lafungin were: A. fumigatus TR34/L98H (n = 25; 0.125, >16,
two. Patients were able to stepdown to oral fluconazole ther­ 0.031 mg/L), A. fumigatus TR46/Y121F/T289A (n = 25; 0.125
apy if deemed appropriate. Preliminary data suggest an 80% >16, 0.063 mg/L), A. flavus (n = 10; 0.063, >16, 0.031 mg/L),
success rate with fosmanogepix with a favorable safety profile. and A. nidulans (n = 10; 0.125, 0.5, 0.063 mg/L) [108].
Several ongoing trials are evaluating fosmanogepix for Olorofim’s activity was maintained in azole-resistant isolates
additional indications (Table 4). The APEX study is an open- regardless of mechanism. Additionally, geometric mean MICs
label, Phase 2 trial for IC and candidemia caused by were lowest for olorofim versus voriconazole (0.26 vs. 5.66 mg/
C. auris [104]. AEGIS is an ongoing Phase 2 study, recruit­ L) for 30 isolates of Lomentospora prolificans [109].
ing patients with IA or other invasive rare mold infections Investigations of inducing resistance in A. fumigatus via serial
(Scedosporium spp., Fusarium spp., and Mucorales) [105]. passage and drug gradients required 40 passages before mod­
Fosmanogepix shows great promise for the treatment of est increases in MIC were observed compared to as low as 10
IFIs, especially MDR fungi including C. auris, with access to with voriconazole, indicating a high barrier to resistance for
a reliable oral option for ease of transition, good tolerabil­ olorofim [9].
ity without expected major DDIs, and penetration to diffi­ Olorofim is currently being evaluated in a Phase 2b clinical
cult sites of infection where few therapeutic options exist. trial (FORMULA-OLS) for patients with resistant IFIs and/or
those with limited therapeutic options (Table 4) [110]. This is
a single-arm, open-labeled trial in patients with IFIs due to
Lomentospora prolificans, Scedosporium, Aspergillus spp., and
3.5. Orotomides other resistant fungi. Olorofim will be dosed according to
Orotomides are a novel class of antifungals with a promising plasma levels to a maximum daily dose of 300 mg, possibly
agent in development, olorofim (F901318), which reversibly indicating a role for TDM. The primary outcome will evaluate
targets the enzyme dihydroorotate dehydrogenase (DHODH). overall clinical and mycological responses at day 42.
This mechanism inhibits the biosynthesis of pyrimidine Outside this clinical trial, there are only case reports doc­
thereby preventing fungal growth. The high degree of speci­ umenting success in disseminated lomentosporiosis and coc­
ficity and activity makes this a promising antifungal class for cidioidomycosis in humans [107]. Due to its niche spectrum of
resistant IFIs. activity, this agent will likely be reserved for identified or
strongly suspected mold infections, particularly those refrac­
tory to currently available options. In empiric situations, com­
3.5.1. Olorofim bination with other therapies may be warranted to preserve
Olorofim’s activity is lacking against Mucorales, Candida, and spectrum against yeasts and Mucorales until microbiological
Cryptococcus spp.; however, it has great potency against confirmation can be obtained.
Aspergillus spp. including strains with reduced azole suscept­
ibility, and other difficult-to-treat molds such as Lomentospora
prolificans and Scedosporium spp [98,106]. Currently, both oral 4. Conclusion
and IV olorofim are in Phase 2 trials for Aspergillus and difficult Current therapeutic options for IFIs are limited by variable PK,
molds in patients with refractory disease, resistance, or intol­ toxicity, DDIs, and emerging resistance. Promising novel anti­
erance to available agents. FDA breakthrough designation, fungals are now being developed which offer extended spec­
Qualified Infectious Disease Product status, and orphan drug trums of activity (Table 3), greater potency, more convenient
designation have been granted for olorofim in the manage­ dosing formulations, and perhaps better tolerability with
ment of several invasive mold infections and coccidioidomy­ fewer significant DDIs (Table 2). In cases of resistance or
cosis refractory to or otherwise unable to be treated with difficult-to-treat IFIs, this pipeline may offer a reprieve from
SOC [67]. clinical dilemmas currently faced by patients and providers. As
Olorofim has a large volume of distribution (3 L/kg) and IFIs continue to impose a significant morbidity and mortality,
a potentially therapeutic CNS distribution with repeat dosing we must optimize our currently available agents even as
in mice [106]. Olorofim is >2200-fold more potent against additional agents become available.
A. fumigatus DHODH compared to the recombinant human
enzyme, suggesting that it has reasonable fungal specific
mechanisms and an expected favorable safety profile [9]. For 5. Expert opinion on novel antifungals
healthy volunteers, multiple dosing studies showed no clini­ Not since the 1990s has there been a pipeline ready to change
cally significant changes in vital signs, electrocardiograms, or the landscape of antifungals and the treatment of IFIs. The role
laboratory variables observed after 5 days. It has good bioa­ of these agents and best practices for each IFI will evolve
vailability (>45%) allowing for the development of both oral greatly in the years to come. We offer a framework and
and IV formulations with a half-life of approximately potential roles in therapy for each of these agents to highlight
20–30 hours [67]. Olorofim is an inhibitor of CYP3A4 and how transformative this age of new antifungals may be, even
10 S. W. MUELLER ET AL.

Table 2. Novel antifungals [1,61,67,69].


Class Mechanism of Action Agents Advantages Disadvantages Potential roles
Tetrazoles Inhibit lanosterol 14α- Oteseconazole ● Minimal DDIs ● Spectrum of activity ● RVVC
demethylase (CYP51) ● Oral ● Onychomycosis
● Prolonged t1/2
VT-1129 ● Minimal DDIs ● Targeted spectrum of ● Cryptococcal meningitis
● Oral activity
● Prolonged t1/2
VT-1598 ● Minimal DDIs ● Higher in vitro MICs ● Potential for MDR infections
● Broad spectrum observed for triazole (C. auris)
of activity resistant A. fumigatus ● Coccidioidomycosis
● Prolonged t1/2
Glucan Synthesis Inhibit 1,3-βD-glucan Rezafungin ● Once weekly ● No oral formulation ● Treatment or prophylaxis options
Inhibitors synthesis dosing ● Lacks activity: for IFIs
(prolonged t1/2) Mucorales, Fusarium ● Potential for MDR infections
● Minimal DDIs spp. (C. auris)
● Minimal AEs (no ● Limited CNS penetra­ ● Improved adherence
hepatoxicity com­ tion ● May retain in vitro activity against
pared to other some caspofungin resistant Candida,
echinocandins) unknown clinical relevance
● Extensive tissue
penetration and
distribution
Ibrexafungerp ● Oral and IV ● Lacks activity: ● Alternative in resistant or intolerant
● Minimal DDIs Mucorales, Fusarium therapies
● Minimal AEs spp. ● May retain in vitro activity against
● Extensive tissue ● Limited CNS penetra­ echinocandin resistant isolates
penetration and tion ● Oral/IV formulation may become
distribution first-line option for IC as initial or
● Broad spectrum stepdown therapy for in IC, IA, VVC,
of activity other refractory IFIs
Polyene Inhibit ergosterol Encochleated ● Oral ● Gastrointestinal AEs ● May not supersede IV therapy in
binding to fungal Amphotericin ● Less toxicity com­ ● Cost clinically unstable
membrane B pared to IV ● Stability ● Potential role in consolidation of
formulation therapy
● Minimal DDIs
Glycosyl- Inhibit Gwt1, GPI anchor Fosmanogepix ● Oral and IV ● No C. krusei coverage ● Potential for first-line treatment in
phosphatidylinositol protein synthesis ● Broad-spectrum ● Mixed Mucorales IFIs
inhibitor of activity activity ● Potential for MDR and difficult-to-
● Once daily dosing treat infections
● Minimal DDIs (No
significant
CYP3A4
inhibition)
● Minimal AEs
● Extensive tissue
penetration and
distribution
● Potential CNS
penetration
Orotomide Inhibit DHODH synthesis Olorofim ● Oral and IV ● Lacks activity: ● Mold targeted or salvage therapy
● Broad-spectrum Mucorales, Candida ● Lomentospora prolificans
of activity Cryptococcus spp. ● CNS coccidioidomycosis, but may
● Potential CNS ● Undergoes CYP3A4 require repetitive dosing and TDM
penetration metabolism/weak
inhibitor of CYP3A4,
potential for DDIs

AE: adverse events; CNS: central nervous system; DDI: drug–drug interactions; DHODH: dihydroorotate dehydrogenase; IA: invasive aspergillosis; IC: invasive
candidiasis; IFIs: invasive fungal infections; IV: intravenous; MDR: multi-drug-resistant; MIC: minimum inhibitory concentration; RVVC: recurrent vulvovaginal
candidiasis; spp: species; t1/2: half-life; VVC: vulvovaginal candidiasis; TDM: therapeutic drug monitoring.

if initial and empiric therapy for IC and IA remain unchanged VT-1598 offers a wider range of activity compared to oteseco­
(Figures 1 and 2). nazole and VT-1129. This agent has superior in vitro activity
against resistant strains, such as C. auris, potentially broadening
its use as a first-line therapy for resistant yeast and mold infec­
5.1. Tetrazoles tions pending clinical efficacy and safety data. VT-1129 is an
Tetrazoles offer many potential advantages over triazoles, exciting future option for cryptococcal meningitis, especially for
namely safety, tolerability, spectrum, potency and fewer DDIs. fluconazole-resistant isolates. Tetrazoles will also be an attractive
EXPERT OPINION ON PHARMACOTHERAPY 11

Table 3. Novel antifungal spectrum of activity [1,61,67,69,98].


Tetrazoles Glucan Synthesis Inhibitors Polyenes GIP Ortomides
Organism Oteseconazole VT-1129 VT-1598 Ibrexafungerp Rezafungin CAmB Fosmanogepix Olorofim
Candida spp. + + + + + +a +b -
C. auris + + + +c + -
C. glabrata + + + + + + + -
Cryptococcus spp. + + + + -
Pneumocystis spp. + +
Aspergillus spp. + + + +d + +
A. fumigatus + + + + + +
Rhizopus spp. + + - - +c ± -
Mucor spp. - - +c ± -
Fusarium spp. - - ±c + +
Lomentospora prolificans ± -c + +
Scedosporium spp. ± ±c + +
Scoulariopsis spp. +
Paecilomyces spp. + +
Trichophyton spp. +
Histoplasma spp. + +c +
Blastomyces spp. + +c +
Coccidiosis spp. + + +c + +
Talaromyces marneffei. + +c +
(+): has shown activity
(-): lacks activity
(±): has mixed activity or mixed activity in some species
()blank indicates: unknown activity at the time of publication
CAmB listed based on in vivo studies unless otherwise specified
Candida spp. includes C. albicans, C. krusei, C. lusitaniae, C. parapsilosis, and C tropicalis unless otherwise specified
Aspergillus spp. includes A. flavus, A. nidulans, A. niger, and A. terrus unless otherwise specified
a
Excluding Candida lusitaniae
b
Fosmanogepix lacks activity against C. krusei and higher MIC to C. kefyr
c
CAmB activity extrapolated from AmB activity
d
Excluding A. terreus
CAmB: encochleated amphotericin B; GIP: Glycosylphosphatidylinositol inhibitor; spp: species.

prophylaxis option where triazoles would otherwise be indi­ Like other echinocandins, rezafungin is IV only and has
cated. Future superiority or non-inferiority trials and registries limited CNS penetration. Pharmacoeconomic analyses will
comparing tetrazole efficacy and cost-effectiveness to triazoles play an important role in determining the cost-effectiveness
are needed to define their role in therapy. Tetrazoles may even­ and role of rezafungin. By utilizing weekly outpatient infu­
tually replace triazoles in many indications. sions, the economic impact of rezafungin may be favorable
for institutions and patients as this could facilitate rapid hos­
5.2. Glucan synthesis inhibitors pital discharge and reduce unnecessary contact with the
healthcare system.
Rezafungin offers enhanced activity against Candida, Of the emerging glucan synthesis inhibitors, ibrexafungerp
Aspergillus spp., P. jiroveci, with a once weekly administration shows the greatest step-forward as both a treatment and
that may ease transitions of care to an outpatient/home infu­ preventative option in IFIs. Its spectrum of activity remains
sion or accelerate discharge near the end of treatment. This broad, maintaining coverage against echinocandin-resistant
will be an improvement in care for patients requiring pro­ Candida and azole-resistant Aspergillus, but similar to echino­
longed echinocandin therapy in cases of triazole-resistant IC candins, it lacks activity against Mucor. Ibrexafungerp may
(e.g., endocarditis, osteomyelitis, etc.) or when triazoles are address unmet critical needs for resistant pathogens such as
nonpreferred. The front-loading regimen may improve blood C. auris as it maintains excellent in vitro activity even in strains
culture clearance in candidemia, but requires further study. with elevated echinocandin MICs. A fungicidal agent with IV
The role of rezafungin prophylaxis among hematopoietic cell and oral options that has excellent tissue penetration (except
transplant recipients should soon be elucidated with the CNS or urine) is an extremely attractive therapy to facilitate
ReSPECT trial, which is of critical importance given a shift transitions of care in similar situations as discussed for
toward outpatient management, overlapping toxicities, and rezafungin.
significant DDIs between triazoles and current and emerging Ongoing clinical studies (CANDLE, CARES, FURI, and
cancer therapies. Rezafungin may mitigate some limitations of SCYNERGIA) will hopefully soon define ibrexafungerp’s role in
commonly utilized pneumocystis pneumonia and antifungal therapy. FURI has already shown a clinical benefit and an accep­
prophylaxis options in the acute setting. Pending positive table safety profile in patients with refractory IFIs or those intol­
results, clinicians should consider the possibility of driving erant to SOC. Clinicians should interpret these trial results in the
echinocandin resistance and monitor the impact on this context of the difficult-to-treat patient population. We await
class. Nevertheless, rezafungin has excellent activity against results of SCYNERGIA, testing a combination therapy with two
Candida and Aspergillus and may maintain clinical effective­ oral agents (ibrexafungerp plus voriconazole) for pulmonary IA,
ness for treatment of some FKS mutated Candida spp. which holds promise given in vitro synergy.
12

Table 4. Novel Antifungals and Phases of Development [46-49,56-59,62,63,83-88,96,103-105,110]


Class Agent Clinical Phase Indication Intervention Outcome
Tetrazoles Oteseconazole Phase 2 Onychomycosis Oteseconazole 300 mg or 600 mg PO daily x2 weeks, then Clinical and mycological cure: Low and high-dose
once weekly vs placebo oteseconazole vs placebo (13.5%-28.1% vs 23.1%)
Phase 3 (ultraVIOLET) RVVC Part 1: Oteseconazole 150 mg PO daily x 2 days vs Culture-verified recurrence: Oteseconazole vs
S. W. MUELLER ET AL.

fluconazole Part 2: Oteseconazole 150 mg PO once fluconazole (5.1% vs 42.2%; p < 0.001)
weekly vs placebo
Phase 3 (VIOLET)* RVVC Oteseconazole 150 mg PO daily x7 days, then 150 mg Culture-verified episodes: No results posted, preliminary
Phase 3 (VIOLET)* weekly vs placebo reports suggest primary endpoint met (p < 0.001)
VT-1129 Phase 1 Cryptococcal VT-1129 in vitro and in vivo No results posted
meningitis
VT-1598 Phase 1 Candida auris VT-1598 in vitro and in vivo No results posted
Cryptococcal
neoformans
Coccidioides
Glucan Synthase Rezafungin (CD101) Phase 3 (ReSTORE) IC and candidemia Rezafungin 400 mg on week 1, then 200 mg weekly vs 30-day all-cause mortality and 14-day global cure
Inhibitors caspofungin
Phase 3 (ReSPECT) IFI prevention in Rezafungin 400 mg on week 1, then 200 mg weekly vs 90-day fungal-free survival
blood and marrow fluconazole 400 mg daily
transplant patients
Ibrexafungerp (SCY-078) Phase 2 (SCYNERGIA) IA Ibrexafungerp 500 mg PO twice daily x2 days, then 500 Primary safety outcome: SAE, discontinuation due to
mg daily in combination with voriconazole vs SAE, death Secondary outcome: global response at
voriconazole monotherapy end of treatment, day 42, and day 84
Phase 3 (FURI) Refractory IFIs Ibrexafungerp 750 mg PO twice daily x2 days, then 750 180-day global response:Interim analysis n=43 with
mg daily; single arm study complete/partial response (56%; n=23) or stable
(27%; n=11)
Phase 3 (CANDLE) RVVC Ibrexafungerp 300 mg PO twice daily for 4 weeks vs 24-week clinical success: No results
placebo
Phase 3 (CARES) Refractory IFIs Ibrexafungerp 750 mg PO twice daily x2 days, then 750 90-day global response: No results (2 cases reported
mg daily; single arm study successful)
Polyenes Encochleated AmB (CAmB; Phase 2 (EnACT) Cryptococcal CAmB dose-escalating study vs IV AmB + 5FC “standard of Reduction in fungal cell counts: no results posted
MAT2203) meningitis care”
Glycosylphos- Fosmanogepix (APX001) Phase 2 (SURGE) IC and candidemia Fosmanoepix 1000 mg IV twice daily x1 day, then 600 mg Treatment success at the end of treatment or 49 days:
phatidylinositol IV once daily, with option to switch to 700 mg PO daily 80% success and 86% survival rates. No treatment
Inhibitors after day 3; single arm study related SAEs or discontinuation
Phase 2 (APEX) Candida auris Fosmanoepix 1000 mg IV twice daily x1 day, then 600 mg Treatment success at the end of treatment or 42 days:
IV daily, with option to switch to 800 mg PO daily after No results posted
day 3; single arm study
Phase 2 (AEGIS) IA and rare molds Fosmanoepix IV load followed by IV/PO (dosing All-cause mortality at day 42: no results posted
unreported); single arm
Orotomides Olorofim (F901318) Phase 2 (FORMULA- IFIs including IA, rare Olorofim PO titrated to plasma levels, maximum 300 mg/ Clinical, mycological and radiological response at day 42:
OLS) and difficult-to- day; single arm no results posted
treat molds
AmB: Amphotericin B; BTD: Breakthrough designation; IA: Invasive Aspergillosis; IC: Invasive Candidiasis; IV: Intravenous; QIDP: Qualified Infectious Disease Product; IFI: Invasive Fungal Infection; PO: Oral; RVVC: Recurrent
Vulvovaginal Candidiasis; SAEs: Study Adverse Events; 5FC: Flucytosine*Oteseconazole phase 3 VIOLET are two global trials with identical study designs
EXPERT OPINION ON PHARMACOTHERAPY 13

Additionally, ibrexafungerp’s use in prophylaxis of intrinsically resistant molds that result in dismal out­
Pneumocystis, IC, and IA is promising, as currently used agents comes [110].
carry a greater risk of DDIs, toxicities, and resistance in compar­ Antifungal development has provided us with some
ison to early ibrexafungerp data. This may translate to overall improved options for IFIs that we continue to optimize in
better outcomes in patient quality of life, safety, and cost. practice to date. Coupled with better diagnostics and com­
Ibrexafungerp is fast-tracked for VVC, but has already proven parative clinical studies, patient outcomes have improved.
useful for step-down therapy in IC and for refractory IFIs. With an emerging armamentarium of antifungals with unique
mechanisms of action and substantial improvements on famil­
iar mechanisms, we anticipate patient outcomes will continue
5.3. Polyenes
to improve with appropriate use of these agents. We can have
CAmB is a potent and broad-spectrum oral antifungal activity confidence that the cavalry is coming.
against Candida, Aspergillus, and Cryptococcus spp (presumably
all AmB susceptible fungus). CAmB has fewer side effects and
increased convenience compared to IV AmB formulations. An List of abbreviations
oral regimen offers a practical approach for prophylaxis in
AmB: Amphotericin B
patients with hematological malignancies and for outpatient ARAF: Azole-resistant A. fumigatus
treatment of cryptococcal meningitis. However, based on its AUC: Area under curve
encochleated formulation, there are concerns regarding the CAmB: Encochleated amphotericin B
cost, chemical stability, and tolerability. If nephrotoxicity is CNS: Central nervous system
minimized with CAmB, studies confirming bioavailability in DDIs: Drug-drug interactions
those with expected poor absorption (e.g., critically ill) are DHODH: Dihydroorotate dehydrogenase
needed. CAmB appears well suited as a consolidation regimen EUCAST: European Committee on Antimicrobial Susceptibility
in clinically improving patients or as initial therapy for other­ Testing
wise stable patients that would benefit from AmB at a much FDA: Food and Drug Administration
lower toxicity profile (e.g., Cryptococcus, certain endemic IFIs, GI: Gastrointestinal
GPI: Glycosylphosphatidylinositol
or visceral leishmaniasis). CAmB is being evaluated for crypto­
IA: Invasive aspergillosis
coccal meningitis and could become an initial therapy or IC: Invasive candidiasis
alternative to fluconazole for consolidation, particularly in flu­ ICU: Intensive Care Unit
conazole-resistant strains. Upcoming clinical trials will provide IDSA: Infectious Disease Society of America
additional insight regarding efficacy and safety of IFIs. IFI: Invasive Fungal Infections
IV: Intravenous
MDR: Multi-drug-resistant
5.4. Glycosylphosphatidylinositol (GPI) inhibitors MIC: Minimum inhibitory concentration
PD: Pharmacodynamic
Fosmanogepix has great potential to be used as a first-line PK: Pharmacokinetic
treatment for IFIs. It has broad activity against Candida (includ­ RVVC: Recurrent vulvovaginal candidiasis
ing C. auris), Aspergillus spp., dimorphic fungi, and rare molds SOC: Standard of care
that are often difficult-to-treat, such as Fusarium, Scedosporium TDM: Therapeutic drug monitoring
spp., and some of the Mucorales group. Fosmanogepix has U.S.: United States
excellent bioavailability, allowing for both oral and IV admin­
istration, and is expected to have an acceptable safety profile.
With its broad spectrum of activity and wide tissue distribution Declaration of interest
(including some CNS penetration), fosmanogepix is of great The authors have no other relevant affiliations or financial involvement
appeal for MDR and difficult-to-treat IFIs. with any organization or entity with a financial interest in or financial
conflict with the subject matter or materials discussed in the manuscript
apart from those disclosed.
5.5. Orotomides
Olorofim is a novel antifungal with excellent activity against
Reviewer Disclosures
a broad range of rare or resistant molds and dimorphic fungi.
This includes azole-resistant aspergillosis, endemic mycoses, Peer reviewers on this manuscript have no relevant financial or other
and other rare molds with limited antifungal treatments, relationships to disclose.
such as L. prolificans, Scedosporium, and Scopulariopsis spp.;
however, lacks activity against Candida, Cryptococcus, and Funding
Mucorales. Olorofim may be best suited for cases of confirmed
or suspected rare or resistant pathogens where it retains The authors are supported by a National Institutes of Health (NIH)/
National Center for Advancing Translational Sciences (NCATS) Colorado
activity or as salvage treatment in those unresponsive to
CTSA [Grant Number UL1 TR002535]. Its contents are the authors' sole
therapy. This novel antifungal agent allows for IV or oral responsibility and do not necessarily represent official NIH views. The
administration with limited toxicities and mild CYP3A4 DDIs, funding source was not involved in manuscript writing or in the decision
which may prove to be helpful with rarer pathogens and to submit the article for publication.
14 S. W. MUELLER ET AL.

ORCID 17. Wheat LJ, Freifeld AG, Kleiman MB, et al. Clinical practice guidelines
for the management of patients with histoplasmosis: 2007 update
Scott W. Mueller http://orcid.org/0000-0003-1238-8199 by the infectious diseases Society of America. Clin Infect Dis.
2007;45:807–825.
18. Galgiani JN, Ampel NM, Blair JE, et al. Executive summary: 2016
References Infectious Diseases Society of America (IDSA) clinical practice
guideline for the treatment of coccidioidomycosis. Clin Infect Dis.
1. Rauseo AM, Coler-Reilly A, Larson L, et al. Hope on the horizon: 2016;63:717–722.
novel fungal treatments in development. Open Forum Infectious 19. Nivoix Y, Ledoux M-P, Herbrecht R. Antifungal therapy: new and
Diseases. 2020;7:ofaa016. evolving therapies. Semin Respir Crit Care Med. 2020;41:158–174.
2. Vincent J-L, Rello J, Marshall J, et al. International study of the DOI: 10.1055/s-0039-3400291.
prevalence and outcomes of infection in intensive care units. 20. Ashley ESD, Lewis R, Lewis JS, et al. Pharmacology of systemic
JAMA. 2009;302:2323. antifungal agents. Clin Infect Dis. 2006;43:S28–S39.
3. Donnelly JP, Chen SC, Kauffman CA, et al. Revision and update of 21. Magill SS, Edwards JR, Bamberg W, et al. Multistate point-
the consensus definitions of invasive fungal disease from the prevalence survey of health care–associated infections. N Engl
European Organization for Research and Treatment of Cancer and J Med. 2014;370:1198–1208.
the Mycoses Study Group Education and Research Consortium. Clin 22. Centers for Disease Control and Prevention (U.S.). Antibiotic resis­
Infect Dis. 2020;71:1367-1376 https://doi.org/10.1093/cid/ciz1008. tance threats in the United States, 2019 [Internet]. Centers for
● An updated consensus definition of IFIs including Disease Control and Prevention (U.S.); [cited 2020 Nov 21].
diagnostics. Available from: https://stacks.cdc.gov/view/cdc/82532.
4. Delaloye J, Calandra T. Invasive candidiasis as a cause of sepsis in 23. Novak AR, Bradley ME, Kiser TH, et al. Azole-resistant Aspergillus
the critically ill patient. Virulence. 2014;5:161–169. and echinocandin-resistant candida: what are the treatment
5. Bassetti M, Righi E, Montravers P, et al. What has changed in the options? Curr Fungal Infect Rep. 2020;14:141–152.
treatment of invasive candidiasis? A look at the past 10 years and 24. Miller MA, Lee YM. Applying pharmacogenomics to antifungal
ahead. J Antimicrob Chemother. 2018;73:i14–i25. selection and dosing: are we there yet? Curr Fungal Infect Rep.
6. Statistics Invasive candidiasis, candidiasis, types of diseases Fungal 2020;14:63–75.
Diseases | CDC [Internet]. 2020 [cited 2020 Nov 21]. Available from: 25. Mahmoud G, Long Lisa L, Emily L. Susceptibility of recent Candida
https://www.cdc.gov/fungal/diseases/candidiasis/invasive/statis auris isolates to the novel echinocandin CD101 and comparator
tics.html. antifungal agents. ECCMID; 2017.
7. Schauwvlieghe AFAD, Rijnders BJA, Philips N, et al. Invasive asper­ 26. Geddes-mcalister J, Shapiro RS. New pathogens, new tricks: emer­
gillosis in patients admitted to the intensive care unit with severe ging, drug-resistant fungal pathogens and future prospects for
influenza: a retrospective cohort study. Lancet Respir Med. antifungal therapeutics. Ann N Y Acad Sci. 2019;1435:57–78.
2018;6:782–792. 27. Lockhart SR, Etienne KA, Vallabhaneni S, et al. Simultaneous emer­
8. Taccone F, Van Den Abeele A-M, Bulpa P, et al. Epidemiology of gence of multidrug-resistant Candida auris on 3 continents con­
invasive aspergillosis in critically ill patients: clinical presentation, firmed by whole-genome sequencing and epidemiological
underlying conditions, and outcomes. Crit Care. 2015;19:7. analyses. Clin Infect Dis. 2017;64:134–140.
9. Oliver JD, Sibley GEM, Beckmann N, et al. F901318 represents 28. Livengood SJ, Drew RH, Perfect JR. Combination therapy for inva­
a novel class of antifungal drug that inhibits dihydroorotate sive fungal infections. Curr Fungal Infect Rep. 2020;14:40–49.
dehydrogenase. Proc Natl Acad Sci USA. 2016;113:12809–12814. 29. Arensman K, Miller J, Chiang A, et al. Clinical outcomes of patients
10. Van Der Linden JWM, Snelders E, Kampinga GA, et al. Clinical treated for candida auris infections in a multisite health system,
implications of azole resistance in aspergillus fumigatus, the Illinois, USA. Emerg Infect Dis. 2020;26:876–880.
Netherlands, 2007–2009. Emerg Infect Dis. 2011;17:1846–1854. 30. Herbrecht R, Dw D, Tf P, et al. Voriconazole versus amphotericin
11. Van Matre ET, Evans SL, Mueller SW, et al. Comparative evaluation B for primary therapy of invasive aspergillosis. N Engl J Med.
of isavuconazonium sulfate, voriconazole, and posaconazole for 2002;347:408–415.
the management of invasive fungal infections in an academic 31. Marr KA, Schlamm HT, Herbrecht R, et al. Combination antifungal
medical center. Ann Clin Microbiol Antimicrob. 2019;18:13. therapy for invasive aspergillosis. Ann Intern Med. 2015;162:81–89.
12. Pappas PG, Kauffman CA, Andes DR, et al. Clinical practice guide­ 32. Antifungal resistance in Aspergillus, fungal diseases, CDC [Internet].
line for the management of candidiasis: 2016 update by the infec­ 2020 [cited 2020 Nov 22]. Available from: https://www.cdc.gov/
tious diseases Society of America. Clin Infect Dis. 2016;62:e1–e50. fungal/diseases/aspergillosis/antifungal-resistant.html.
● IDSA clinical practice guideline discussing current manage­ 33. Lestrade PPA, Buil JB, Van Der Beek MT, et al. Paradoxal trends in
ment strategies for IC. azole-resistant Aspergillus fumigatus in a national multicenter sur­
13. Cornely OA, Bassetti M, Calandra T, et al. ESCMID guideline for the veillance program, the Netherlands, 2013–2018. Emerg Infect Dis.
diagnosis and management of Candida diseases 2012: 2020;26:1447–1455.
non-neutropenic adult patients. Clin Microbiol Infect. 34. Vahedi-Shahandashti R, Lass-Flörl C. Novel antifungal agents and
2012;18:19–37. their activity against Aspergillus species. J Fungi. 2020;6:213.
14. Patterson TF, Thompson GR, Denning DW, et al. Practice guidelines 35. Gonçalves SS, Souza ACR, Chowdhary A, et al. Epidemiology and
for the diagnosis and management of aspergillosis: 2016 update by molecular mechanisms of antifungal resistance in Candida and
the infectious diseases society of America. Clin Infect Dis. 2016;63: Aspergillus. Mycoses. 2016;59:198–219.
e1–e60. 36. Rybak JM, Fortwendel JR, Rogers PD. Emerging threat of
● IDSA clinical practice guideline discussing current manage­ triazole-resistant Aspergillus fumigatus. J Antimicrob Chemother.
ment strategies for IA. 2019;74:835–842.
15. Ullmann AJ, Aguado JM, Arikan-Akdagli S, et al. Diagnosis and 37. Yi WM, Schoeppler KE, Jaeger J, et al. Voriconazole and posacona­
management of Aspergillus diseases: executive summary of the zole therapeutic drug monitoring: a retrospective study. Ann Clin
2017 ESCMID-ECMM-ERS guideline. Clin Microbiol Infect. 2018;24: Microbiol Antimicrob. 2017;16:60.
e1–e38. 38. Lepak A, Andes D. Fungal sepsis: optimizing antifungal therapy in
●● ESCMID clinical practice guideline discussing management the critical care setting. Crit Care Clin. 2011;27:123–147.
strategies for IA. 39. Erasmus Medical Center. PCR based detection of azole resistance in
16. Chapman SW, Dismukes WE, Proia LA, et al. Clinical practice guide­ a. fumigatus to improve patient outcome. (azorMan) [Internet].
lines for the management of blastomycosis: 2008 update by the clinicaltrials.gov; [cited 2020 Nov 18]. Report No.: NCT03121235.
infectious diseases society of America. Clin Infect Dis. Available from: https://clinicaltrials.gov/ct2/show/NCT03121235.
2008;46:1801–1812.
EXPERT OPINION ON PHARMACOTHERAPY 15

40. Lepak AJ, Marchillo K, VanHecker J, et al. Posaconazole pharmaco­ mucocutaneous candidiasis. Open Forum Infect Dis. 2016;3
dynamic target determination against wild-type and Cyp51 mutant (S1):1635. https://doi.org/10.1093/ofid/ofw172.1335
isolates of Aspergillus fumigatus in an in vivo model of invasive 55. Schell WA, Jones AM, Garvey EP, et al. Fungal CYP51 inhibitors
pulmonary Aspergillosis. Antimicrob Agents Chemother. VT-1161 and VT-1129 exhibit strong in vitro activity against can­
2013;57:579–585. dida glabrata and C. krusei isolates clinically resistant to azole and
41. Chen L, Krekels EHJ, Verweij PE, et al. Pharmacokinetics and phar­ echinocandin antifungal compounds. Antimicrob Agents
macodynamics of posaconazole. Drugs. 2020;80:671–695. Chemother. 2017;61:e01817–16.
42. Schauwvlieghe AFAD, Buil JB, Verweij PE, et al. High-dose posaco­ 56. Nielsen K, Vedula P, Smith KD, et al. Activity of VT-1129 against
nazole for azole-resistant aspergillosis and other difficult-to-treat Cryptococcus neoformans clinical isolates with high fluconazole
mould infections. Mycoses. 2020;63:122–130. MICs. Med Mycol. 2017;55:453–456.
43. Schauwvlieghe A. Risk factors, diagnosis and management of 57. Wiederhold NP, Xu X, Wang A, et al. In vivo efficacy of VT-1129
(azole-resistant) invasive aspergillosis. 2020. against experimental cryptococcal meningitis with the use of
44. Buil JB, Brüggemann RJM, Wasmann RE, et al. Isavuconazole sus­ a loading dose-maintenance dose administration strategy.
ceptibility of clinical Aspergillus fumigatus isolates and feasibility of Antimicrob Agents Chemother. 2018;62:e01315–18.
isavuconazole dose escalation to treat isolates with elevated MICs. ● Animal model activity of VT-1129 against cryptococcal
J Antimicrob Chemother. 2018;73:263. meningitis.
45. Warrilow AGS, Hull CM, Parker JE, et al. The clinical candidate 58. Wiederhold NP, Patterson HP, Tran BH, et al. Fungal-specific Cyp51
VT-1161 is a highly potent inhibitor of Candida albicans CYP51 inhibitor VT-1598 demonstrates in vitro activity against Candida
but fails to bind the human enzyme. Antimicrob Agents and Cryptococcus species, endemic fungi, including Coccidioides
Chemother. 2014;58:7121–7127. species, Aspergillus species and Rhizopus arrhizus. J Antimicrob
46. Viamet. A phase 2, randomized, double-blind, placebo-controlled, Chemother. 2018;73:404–408.
dose-ranging study to evaluate the efficacy and safety of VT-1161 ●● In vitro activity of VT-1598 demonstrating potential for
oral tablets in the treatment of patients with distal lateral subun­ broad activity as a tetrazole but potential for triazole cross-
gual onychomycosis of the toenail [internet]. clinicaltrials.gov; 2020 resistance.
[cited 2020 Nov 19]. Report No.: NCT02267356. Available from: 59. Wiederhold NP, Lockhart SR, Najvar LK, et al. The fungal
https://clinicaltrials.gov/ct2/show/NCT02267356. Cyp51-specific inhibitor VT-1598 demonstrates in vitro and in vivo
47. Mycovia Pharmaceuticals Inc. A phase 3, randomized, double-blind, activity against Candida auris. Antimicrob Agents Chemother.
placebo-controlled study to evaluate the efficacy and safety of 2019;63:e02233–18.
VT-1161 oral capsules in the treatment of subjects with recurrent ● Demonstrates VT-1598 as active against many C. auris
vulvovaginal candidiasis [internet]. clinicaltrials.gov; 2020 [cited isolates.
2020 Nov 19]. Report No.: NCT03562156. Available from: https:// 60. Chatelon J, Cortegiani A, Hammad E, et al. Choosing the right
clinicaltrials.gov/ct2/show/NCT03562156. antifungal agent in ICU patients. Adv Ther. 2019;36:3308–3320.
● One of various trials anticipated to be crucial for the 61. Pound MW, Townsend ML, Dimondi V, et al. Overview of treat­
approval of oteseconazole. ment options for invasive fungal infections. Med Mycol.
48. Mycovia Pharmaceuticals Inc. A phase 3, randomized, double-blind, 2011;49:561–580.
placebo-controlled study to evaluate the efficacy and safety of
62. Cidara Therapeutics Inc. A phase 3, multicenter, randomized,
VT-1161 oral capsules in the treatment of subjects with recurrent double-blind study of the efficacy and safety of rezafungin for
vulvovaginal candidiasis [internet]. clinicaltrials.gov; 2020 [cited
injection vs. intravenous caspofungin followed by oral fluconazole
2020 Nov 19]. Report No.: NCT03561701. Available from: https://
step down in the treatment of subjects with Candidemia and/or
clinicaltrials.gov/ct2/show/NCT03561701. Invasive Candidiasis [internet]. clinicaltrials.gov; 2020 [cited 2020
● One of various trials anticipated to be crucial for the
Nov 19]. Report No.: NCT03667690. Available from: https://clinical
approval of oteseconazole. trials.gov/ct2/show/NCT03667690.
49. Mycovia Pharmaceuticals Inc. A phase 3, randomized, double-blind
● Will be a pivotal trial for rezafungin approval.
study to evaluate the efficacy and safety of VT-1161 oral capsules
63. Cidara Therapeutics Inc. A phase 3, multicenter, randomized,
versus fluconazole and placebo in the treatment of acute vulvova­ double-blind study of the efficacy and safety of rezafungin for
ginal candidiasis episodes in subjects with recurrent vulvovaginal
injection versus the standard antimicrobial regimen to prevent
candidiasis [internet]. clinicaltrials.gov; 2020 [cited 2020 Nov 19]. invasive fungal diseases in adults undergoing allogeneic blood
Report No.: NCT03840616. Available from: https://clinicaltrials.gov/ and marrow transplantation (The ReSPECT study) [internet]. clini­
ct2/show/NCT03840616.
caltrials.gov; 2020 [cited 2020 Nov 19]. Report No.: NCT04368559.
● One of various trials anticipated to be crucial for the Available from: https://clinicaltrials.gov/ct2/show/NCT04368559.
approval of oteseconazole.
● Will be a pivotal trial for rezafungin approval and use as a
50. Nishimoto AT, Wiederhold NP, Flowers SA, et al. In vitro activities of
prophylaxis agent.
the novel investigational tetrazoles VT-1161 and VT-1598 com­ 64. Zhao Y, Prideaux B, Nagasaki Y, et al. Unraveling drug penetration
pared to the triazole antifungals against azole-resistant strains
of echinocandin antifungals at the site of infection in an
and clinical isolates of Candida albicans. Antimicrob Agents intra-abdominal abscess model. Antimicrob Agents Chemother.
Chemother. 2019;63:e00341–19.
2017;61:e01009–17.
51. Nishimoto AT, Whaley SG, Wiederhold NP, et al. Impact of the
65. Ong V, James KD, Smith S, et al. Pharmacokinetics of the novel
major Candida glabrata triazole resistance determinants on the
activity of the novel investigational tetrazoles VT-1598 and echinocandin CD101 in multiple animal species. Antimicrob Agents
Chemother. 2017;61:e01626–16.
VT-1161. Antimicrob Agents Chemother. 2019;63:e01304–19.
52. Brand SR, Sobel JD, Nyirjesy P, et al. A randomized phase 2 study of 66. Lakota E, Bader J, Ong V, et al. Pharmacological basis of CD101
VT-1161 for the treatment of acute vulvovaginal Candidiasis. Clin efficacy: exposure shape matters. Antimicrob Agents Chemother.
Infect Dis. 2020;ciaa1204. https://doi.org/10.1093/cid/ciaa1204 2017;61:AAC.00758–17.
53. Brand SR, Degenhardt TP, Person K, et al. A phase 2, randomized, 67. Gintjee TJ, Donnelley MA, Thompson GR. Aspiring antifungals:
double-blind, placebo-controlled, dose-ranging study to evaluate review of current antifungal pipeline developments. J Fungi.
the efficacy and safety of orally administered VT-1161 in the treat­ 2020;6:28. DOI:10.3390/jof6010028
ment of recurrent vulvovaginal candidiasis. Am J Obstet Gynecol. 68. Pfaller M, Carvalhaes C, Messer S, et al. Activity of a long-acting
2018;218:624.e1-624.e9. echinocandin, rezafungin, and comparator antifungal agents tested
54. Desai J, Break T, Natarajan M, et al. VT-1129 and VT-1161 have against contemporary invasive fungal isolates: SENTRY program
in vitro activity against Candida isolates from patients with chronic 2016-2018. Antimicrobial Agents and Chemotherapy. 2020;64.
16 S. W. MUELLER ET AL.

69. Van Daele R, Spriet I, Wauters J, et al. Antifungal drugs: what brings refractory to or intolerant of standard antifungal treatment (FURI)
the future? Med Mycol. 2019;57:S328–S343. [internet]. clinicaltrials.gov; 2020 [cited 2020 Nov 19]. Report No.:
70. Thompson GR, Soriano A, Skoutelis A, et al. Rezafungin versus NCT03059992. Available from: https://clinicaltrials.gov/ct2/show/
caspofungin in a phase 2, randomized, double-blind study for the NCT03059992.
treatment of candidemia and invasive candidiasis- the STRIVE trial. ●● Important future role for ibrexafungerp.
Clin Infect Dis. 2020;ciaa1380. https://doi.org/10.1093/cid/ciaa1380 86. Scynexis, Inc. A phase 3, multicenter, randomized, double-blind,
●● Comparative rezafungin to a standard of care regimen for placebo-controlled study to evaluate the efficacy and safety of oral
candidemia and IC. ibrexafungerp (SCY-078) compared to placebo in subjects with
71. Cushion MT, Ashbaugh A, Ong V. Rezafungin prevention of pneu­ recurrent vulvovaginal candidiasis (VVC) [Internet]. clinicaltrials.
mocystis pneumonia and pneumocystis reactivation using different gov; 2020 [cited 2020 Nov 19]. Report No.: NCT04029116.
doses and durations of prophylaxis in a mouse model. Blood. Available from: https://clinicaltrials.gov/ct2/show/NCT04029116.
2019;134:3266. 87. Scynexis, Inc. Open-label study to evaluate the efficacy, safety,
72. Barat S, Borroto-Esoda K, Ghannoum M, et al. Activity of tolerability and pharmacokinetics of oral ibrexafungerp (SCY-078)
Ibrexafungerp (Formerly SCY-078) against candida auris: in vitro, as an emergency use treatment for patients with candidiasis,
in vivo, and clinical case studies of candidemia. Open Forum Infect including candidemia, caused by candida auris [internet]. clinical­
Dis. 2019;6:S307. trials.gov; 2020 [cited 2020 Nov 19]. Report No.: NCT03363841.
73. Pfaller MA, Messer SA, Motyl MR, et al. Activity of MK-3118, a new oral Available from: https://clinicaltrials.gov/ct2/show/NCT03363841.
glucan synthase inhibitor, tested against Candida spp. by two inter­ ●● An important future role for ibrexafungerp.
national methods (CLSI and EUCAST). J Antimicrob Chemother. 88. Juneja D, Singh O, Tarai B, et al. Successful treatment of two patients
2013;68:858–863. with Candida auris Candidemia with the investigational agent, oral
74. Pfaller MA, Messer SA, Motyl MR, et al. In vitro activity of a new oral ibrexafungerp (formerly SCY-078), from the CARES study. 2019.
glucan synthase inhibitor (MK-3118) tested against Aspergillus spp. 89. Aigner M, Lass-Flörl C. Encochleated amphotericin b: is the oral
by CLSI and EUCAST broth microdilution methods. Antimicrob availability of amphotericin B finally reached? J Fungi. 2020;6:66.
Agents Chemother. 2013;57:1065–1068. 90. Shende P, Khair R, Gaud RS. Nanostructured cochleates: a
75. Wring S, Murphy G, Atiee G, et al. Clinical pharmacokinetics and multi-layered platform for cellular transportation of therapeutics.
drug-drug interaction potential for coadministered SCY-078, an Drug Dev Ind Pharm. 2019;45:869–881.
oral fungicidal glucan synthase inhibitor, and tacrolimus. Clin 91. Cuddihy G, Wasan EK, Di Y, et al. The development of oral ampho­
Pharmacol Drug Dev. 2019;8:60–69. tericin B to treat systemic fungal and parasitic infections: has the
76. Lima SL, Colombo AL, de Almeida Junior JN. Fungal cell wall: myth been finally realized? Pharmaceutics. 2019;11:99.
emerging antifungals and drug resistance. Front Microbiol. ● An overview of amphotericin b development as an oral
2019;10. agent.
77. Arendrup MC, Jørgensen KM, Hare RK, et al. Vitro activity of ibrex­ 92. Mannino R, Perlin D Oral dosing of encochleated amphotericin
afungerp (SCY-078) against Candida auris isolates as determined by B (CAmB): rapid drug targeting to infected tissues in mice with
EUCAST methodology and comparison with activity against invasive Candidiasis. 2015.
C. albicans and C. glabrata and with the activities of six comparator 93. Delmas G, Park S, Chen ZW, et al. Efficacy of orally delivered
agents. Antimicrob Agents Chemother. 2019;64:e02136–19. cochleates containing amphotericin B in a murine model of
78. Ghannoum M, Arendrup MC, Chaturvedi VP, et al. Ibrexafungerp: aspergillosis. Antimicrob Agents Chemother. 2002;46:2704–2707.
a novel oral triterpenoid antifungal in development for the treat­ 94. Skipper CP, Atukunda M, Stadelman A, et al. Phase I EnACT trial of
ment of candida auris infections. Antibiotics. 2020;9:539. the safety and tolerability of a novel oral formulation of amphoter­
79. Azie N, Angulo D, Dehn B, et al. Oral Ibrexafungerp: an investiga­ icin B. Antimicrob Agents Chemother. 2020;64:e00838–20.
tional agent for the treatment of vulvovaginal candidiasis. Expert 95. Matinas BioPharma Nanotechnologies, Inc. A multi-center rando­
Opin Investig Drugs. 2020;29:893–900. mized study to evaluate the safety, tolerability, and efficacy of oral
80. Berkow EL, Angulo D, Lockhart SR. In vitro activity of a novel glucan encochleated amphotericin B (CAMB/MAT2203) compared with
synthase inhibitor, SCY-078, against clinical isolates of candida oral fluconazole in the treatment of moderate to severe
auris. Antimicrob Agents Chemother. 2017;61:e00435–17. Vulvovaginal Candidiasis (VVC) [Internet]. clinicaltrials.gov; 2018
81. Zhu Y, Barat S, Borroto-Esoda K, et al. In vitro efficacy of novel [cited 2020 Nov 19]. Report No.: NCT02971007. Available from:
glucan synthase inhibitor, ibrexafungerp (SCY-078), against https://clinicaltrials.gov/ct2/show/NCT02971007.
multidrug- and pan-resistant Candida auris isolates from the out­ 96. Matinas BioPharma Nanotechnologies, Inc. Encochleated oral
break in New York. 2019. Amphotericin for Cryptococcal meningitis Trial (EnACT) [Internet].
● In vitro data of ibrexafungerp in an important role of treat­ clinicaltrials.gov; [cited 2020 Nov 22]. Report No.: NCT04031833.
ing C. auris. Available from: https://clinicaltrials.gov/ct2/show/results/
82. Spec A, Pullman J, Thompson GR, et al. MSG-10: a phase 2 study of NCT04031833.
oral ibrexafungerp (SCY-078) following initial echinocandin therapy 97. Shaw KJ, Ibrahim AS. Fosmanogepix: a review of the first-in-class
in non-neutropenic patients with invasive candidiasis. J Antimicrob broad spectrum agent for the treatment of invasive fungal
Chemother. 2019;74:3056–3062. infections. J Fungi. 2020;6:239.
● Viability of ibrexafungerp as a stepdown agent. 98. Perfect JR. The antifungal pipeline: a reality check. Nat Rev Drug
83. Scynexis, Inc. A multicenter, randomized, double-blind study to Discov. 2017;16:603–616.
evaluate the safety and efficacy of the coadministration of 99. Gebremariam T, Alkhazraji S, Alqarihi A, et al. Fosmanogepix
SCY-078 with voriconazole in patients with invasive pulmonary (APX001) is effective in the treatment of pulmonary murine mucor­
aspergillosis [internet]. clinicaltrials.gov; 2020 [cited 2020 Nov 19]. mycosis due to Rhizopus arrhizus. Antimicrob Agents Chemother.
Report No.: NCT03672292. Available from: https://clinicaltrials.gov/ 2020;64:e00178–20.
ct2/show/NCT03672292. 100. Mansbach R, Shaw KJ, Hodges MR, et al. Absorption, distribution,
●● Results from this study could point to a new treatment and excretion of 14C-APX001 after single-dose administration to
regimen in IA. rats and monkeys. Open Forum Infect Dis. 2017;4:S472.
84. SCYNEXIS, Inc. (SCYX) [Internet]. SCYNEXIS, Inc. [cited 2020 Nov 22]. 101. Pfaller MA, Huband MD, Flamm RK, et al. In vitro activity of APX001A
Available from: https://www.scynexis.com. (manogepix) and comparator agents against 1,706 fungal isolates
85. Scynexis, Inc. Open-label study to evaluate the efficacy and safety of collected during an international surveillance program in 2017.
SCY-078 (Ibrexafungerp) in patients with fungal diseases that are Antimicrob Agents Chemother. 2019;63:e00840–19.
EXPERT OPINION ON PHARMACOTHERAPY 17

102. Hager CL, Larkin EL, Long L, et al. In vitro and in vivo evaluation of ● Fosmanogepix clinical use reports.
the antifungal activity of APX001A/APX001 against Candida auris. 106. Wiederhold NP. Review of the novel investigational antifungal
Antimicrob Agents Chemother. 2018;62:e02319–17. olorofim. J Fungi. 2020;6:122.
103. Amplyx Pharmaceuticals. An open-label study to evaluate the efficacy ● Olorofim specific review.
and safety of APX001 in non neutropenic patients with candidemia, 107. Kennedy T, Allen G, Steiner J, et al. An open label study in healthy
with or without invasive candidiasis, inclusive of patients with sus­ volunteers to evaluate the potential for cytochrome P450 3A4
pected resistance to standard of care antifungal treatment [internet]. inhibition by f901318 using oral midazolam as a probe. 2017.
clinicaltrials.gov; 2020 [cited 2020 Nov 19]. Report No.: NCT03604705. 108. Buil JB, Rijs AJMM, Meis JF, et al. In vitro activity of the novel
Available from: https://clinicaltrials.gov/ct2/show/NCT03604705. antifungal compound F901318 against difficult-to-treat
104. Amplyx Pharmaceuticals. An open-label study to evaluate the effi­ Aspergillus isolates. J Antimicrob Chemother. 2017;72:2548–2552.
cacy and safety of APX001 in patients with candidemia and/or 109. Biswas C, Law D, Birch M, et al. In vitro activity of the novel
invasive candidiasis caused by candida auris [internet]. clinical­ antifungal compound F901318 against Australian Scedosporium
trials.gov; 2020 [cited 2020 Nov 19]. Report No.: NCT04148287. and Lomentospora fungi. Medical Mycology. 2018;56:1050-1054.
Available from: https://clinicaltrials.gov/ct2/show/NCT04148287. https://doi.org/10.1093/mmy/myx161
● Fosmanogepix clinical use reports. 110. F2G Ltd. Phase IIb Study of F901318 as Treatment of Invasive
105. Amplyx Pharmaceuticals. A phase 2, open-label study to evaluate Fungal Infections Due to Lomentospora Prolificans,
the safety and efficacy of APX001 in the treatment of patients with Scedosporium Spp., Aspergillus Spp., and Other Resistant Fungi
invasive mold infections caused by Aspergillus species or rare in Patients Lacking Suitable Alternative Treatment Options
molds [internet]. clinicaltrials.gov; 2020 [cited 2020 Nov 19]. [Internet].. clinicaltrials.gov; 2021 [cited 2021 Feb 25]. Report
Report No.: NCT04240886. Available from: https://clinicaltrials.gov/ No.: NCT03583164. Available from: https://clinicaltrials.gov/ct2/
ct2/show/NCT04240886. show/NCT03583164.

You might also like