Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2021 Jun 1.
Published in final edited form as: Curr Fungal Infect Rep. 2020 Mar 26;14(2):141–152. doi: 10.1007/s12281-020-00379-2

Azole-resistant Aspergillus and Echinocandin-resistant Candida – What are the treatment options?

Alison R Novak 1, Mary E Bradley 1, Tyree H Kiser 1, Scott W Mueller 1
PMCID: PMC7375389  NIHMSID: NIHMS1579779  PMID: 32699568

Abstract

Purpose of Review

This review summarizes current treatment options for echinocandin-resistant Candida spp. (ERC) and azole-resistant Aspergillus fumigatus (ARAF), emphasizing recent in vitro/in vivo data, clinical reports, and consensus statements.

Recent Findings

Advances in ERC and ARAF treatment are limited to specific antifungal combinations and dose optimization but remain reliant on amphotericin products. Although novel antifungals may provide breakthroughs in the treatment of resistant fungi, these agents are not yet available. Early identification and appropriate treatment remain a paramount, albeit elusive, task.

Summary

When either ERC or ARAF are suspected or proven, amphotericin products remain the cornerstone of initial therapy. For ERC, azoles are de-escalation options for susceptible isolates in stable patients to avoid amphotericin toxicities. Although combination echinocandin with high-dose salvage posaconazole or isavuconazole may be attempted in ARAF, it requires careful consideration following patient stabilization. Future research defining optimal therapies and early identification of ERC and ARAF is of extreme importance.

Keywords: Antifungal, Resistance, Echinocandin-Resistant Candida, Azole-Resistant Aspergillus, Invasive Fungal Infection, Amphotericin, Posaconazole, Isavuconazole, Voriconazole, Caspofungin, Micafungin, Anidulafungin

Introduction

The incidence of invasive fungal infections due to Candida spp. and Aspergillus spp. continues to increase as the population of immunocompromised patients increases [1, 2, 3, 4••, 5]. Available treatment options remain limited due to the lack of new antifungal class approvals. Mainstay options for the treatment of invasive fungal infections include polyenes, azoles, echinocandins, and flucytosine (5-FC). Although these agents are effective in treating the majority of fungal infections, the lack of advancement in alternative therapies poses a problem when fungal pathogens acquire resistance to preferred agents, which leads to extensive patient morbidity and mortality [2, 3, 4••].

Emerging resistance is anticipated to intensify in the years to come due to a variety of driving factors. Some Candida spp. can breed inherent resistance by changes in the DNA sequencing. Acquired resistance, noted in both Candida spp. and Aspergillus spp., occurs following drug exposure giving the potential for the pathogen to adapt over time [5]. Recent concerning trends of resistance to generally preferred antifungal agents are on the rise, including but not limited to, echinocandin-resistant Candida spp. (ERC) and azole-resistant Aspergillus fumigatus (ARAF) [5].

Much of the literature available describes resistance patterns of ERC and ARAF but lacks robust evidence to support treatment recommendations for these infections. This review will explore the mechanisms of resistance for ERC and ARAF and provide a treatment approaches to combat these invasive infections.

Echinocandin-Resistant Candida

Background/Epidemiology

Candida infections account for the majority of the fungal infections in immunocompromised patients and are recognized as the 4th leading cause of hospital-acquired bloodstream infections in the United States [1, 2, 6, 7]. Among these invasive Candida infections, > 90% are caused by the most common pathogens of C. albicans, C. glabrata, C. tropicalis, C. parapsilosis, and C. krusei. Each species may vary by regional ecology and produce unique virulence factors and resistance patterns, especially to echinocandins (Table 1) [7, 8]. Currently, the IDSA guidelines recommend echinocandins as preferred first-line agents for patients with invasive candidiasis, with recommendations to step down to azole antifungals in patients who are clinically stable within 5–7 days or who have azole-susceptible isolates. Echinocandins are also recommended as options in patients with prior azole exposures [7].

Table 1:

Resistance Profiles for Echinocandin-Resistant Candida and Azole-Resistant Aspergillus

Species Resistance Rates (%) Mechanism of Resistance****
Echinocandin Resistant Candida spp.*
C. albicans 0.0 (A)
<0.1–0.2 (C)
0.0–0.2(M)

FKS1/2 HS1 alterations

FKS1: L662W, S629P, F625S, R631S; S645P, F641S, F641I, S629P, S654P, F650S, F641S, F641L

FKS2: S663P, L662W, F659S/V/Y, D666E/K753Q, F659_del
C. glabrata 1.5–2.8 (A)
1.3–6.9 (C)
1.0–2.8 (M)
C. parapsilosis 0.0–0.4 (A)
0.0 (C)
0.0 (M)
C. tropicalis 0.0–1.3 (A)
0.0–2.0 (C)
0.0–2.0 (M)
C. krusei 0.0–3.6 (A)
0.0–7.3 (C)
0.0 (M)
C. auris** 7–10 (A, M)
Azole Resistant Aspergillus spp. ***
A. fumigatus 0–3.2 Point mutations: G54, G138, G448S, M220

Tandem repeats/Combination mutations: TR34/L98H, TR46/Y121/T289A

Non-CYP mediated mutations:
Efflux pumps: ABC and MFS transporters
HMG-CoA mutations: hmg1, erg6, HMG-CoA point mutations
Non-fumigatus spp. Not extensively documented
*

From SENTRY Antifungal Surveillance program 1997 – 2016 gathered from 135 medical centers in 39 countries [10•].

**

No current clinical breakpoints reported for C. auris. Resistance rates estimated using EUCAST and CLSI Candida spp. breakpoints [15••].

***

Extrapolated from a study evaluating isolates from 22 centers in 19 countries (range: 0–26.1%). A surveillance study published in 2014 reported a 0% incidence of TR34/L98H mutations in 1,026 clinical isolates in the United States, but case reports of clinical case of azole-resistant A. fumigatus in the United States have been published [35•, 3739]

****

not a comprehensive list of point mutations associated with echinocandin-resistant Candida spp. or azole-resistant Aspergillus spp.

A, anidulafungin; C, caspofungin; M, micafungin; HS, hot spot; ABC, ATP-binding cassette; MFS, major facilitator superfamily

Echinocandins act as fungicidal agents to yeast by inhibiting the biosynthesis of glucan polymers specifically at 1,3-β-d-glucan synthase, an enzyme not found in mammalian cells [2, 5, 8]. The enzyme consists of two subunits: Rho1p, which is a guanosine triphosphate binding protein, and FKS, which is the catalytic component further classified into FKS1, FKS2, and FKS3 [5, 9]. Table 1 outlines the echinocandin resistance patterns of common Candida spp. adopted from 20 years of SENTRY data evaluating 15,308 isolates worldwide [10•]. Although echinocandin-resistant Candida (ERC) prevalence is low overall at this time, C. glabrata, C. krusei, and C. auris have the highest probability of resistance [5, 10•]. Comparatively, C. parapsilosis and C. guilliermondii exhibit the highest intrinsic echinocandin MICs [1, 10•].

Notably, the United States Centers for Disease Control and Prevention (CDC) and Public Health England (PHE) have issued recent warnings about the emergence of C. auris, a multidrug-resistant Candida spp. It is imperative to quickly identify C. auris in the clinical setting due to its vast ability to cause outbreaks; however, it is often misidentified leading to inappropriate management [8, 11, 12]. Rapid genotyping has been recommended in patients who are infected or colonized with C. auris to detect for ERC genes to help guide therapy [13]. Although C. auris is usually susceptible to echinocandins in vitro, there have been concerning cases of high-level echinocandin resistance in the clinical setting with resistance rates of up to 10% based on European Committee on Antimicrobial Susceptibility Testing (EUCAST)/Clinical Laboratory and Standards Institute (CLSI) methodology (Table 1) [14, 15••].

Mechanisms of Resistance

Two main mechanisms have been reported among Candida spp.: intrinsic point mutations and adaptive stress response [1]. The main resistance mechanism described in the literature relates to point mutations, specifically in FKS1 or FKS2 genes on highly selective “hot spot” regions, reducing susceptibility to echinocandins by up to 3000-fold [1, 2, 16, 17, 18]. However, it is apparent that not all FKS mutations are predictive of the degree of echinocandin resistance [10•, 18, 19]. As described above, these mutations can occur intrinsically in some Candida spp. (e.g., C. parapsilosis increasing MIC) or adaptively, which often occurs after being exposed to echinocandins over time.

Even C. albicans, normally susceptible to many antifungals, can develop resistance to echinocandins when exposed over a long period of time. C. albicans clinical isolates exposed to ~ 6 weeks of caspofungin and anidulafungin developed echinocandin resistance. ERC was conferred by a mutation in FKS1 gene leading to amino acid change S645P. This study also noted that the acquired resistance may change fitness and virulence of the organism in an insect model [20].

A series of clinical isolates of C. kefyr from patients with hematologic malignancies that displayed reduced echinocandin susceptibilities were assessed for mechanisms of resistance. The isolates did not display genetic relatedness but did highlight that FKS1 mutations may result in differential echinocandin sensitivities. Specifically, even though the S645P mutation showed resistance to anidulafungin, micafungin, and caspofungin, caspofungin retained some activity (MIC = 2 mg/L) compared to anidulafungin or micafungin (each MIC > 8 mg/L) [21].

Although the majority of resistance is associated with FKS mutations, not all ERC is directly linked to FKS mutations in the hot spot region. A study screened 1380 C. glabrata clinical isolates and assessed resistance mechanisms of 77 echinocandin-resistant isolates. Investigators were able to identify FKS mutations in 51 isolates. Some C. glabrata isolates considered ERC by MIC did not have FKS mutations identified. Investigators observed that micafungin MIC values in candidemia were the strongest predictor of isolates possessing FKS mutations, with 91% expressing MICs in the resistant range. Of these 1380 C. glabrata strains, 10.3% were fluconazole resistant [17]. Alarmingly, 36.2% of echinocandin-resistant C. glabrata strains were resistant to fluconazole. This multidrug resistance relationship has been noted in other studies as well [6, 22, 23]. The cause of the co-resistance in isolates is unknown, due to the azoles and echinocandins having differences in their mechanisms of action and resistance patterns. One proposed mechanism has been reported to be a disrupted MSH2 mismatch repair gene that can potentially produce multidrug-resistant strains, although still often susceptible to amphotericin (AMB) [22]. Alternatively, this co-resistance may be a result of decades of selective pressure.

Current Treatment Options

Suspicion of ERC should arise if clinical worsening or breakthrough Candida infection occurs despite appropriate source control and dosing of the echinocandin or if an FKS mutation is detected in the isolate. Risk factors associated with ERC include hospitalization in the previous 90 days, receipt of total parenteral nutrition in the previous 14 days, a previous episode of candidemia, prior echinocandin exposure, and known colonization with fluconazole-resistant Candida spp. [9]. These factors, including local susceptibility patterns and species-specific risk (i.e., C. glabrata and C. auris more likely than C. albicans), should be considered in the level of suspicion for ERC. Beyond standard phenotypic susceptibility testing, novel methods (e.g., molecular detection of FKS mutations) to identify resistance may be validated in the future. When ERC is suspected, a change of therapy while awaiting culture and susceptibility results may provide earlier optimal antifungal therapy (Table 2) [8, 10•].

Table 2:

Treatment Options for Echinocandin-resistant Candida spp. and Azole-resistant Aspergillus spp.

Antifungal Strategy Site of Infection Patient Population Comments
Echinocandin Resistant Candida spp.
First-line therapy
Lipid Formulation Amphotericin-B Monotherapy All sites Clinically Stable, Clinically Unstable Recommended first-line treatment for echinocandin-resistant Candida spp. Amphotericin products can be used in combination with other agents based on site of infection, but should serve as the backbone for therapeutic plans.
Recommended Lipid Formulation Amphotericin-B dosing strategy is 3–5 mg/kg/day.
Amphotericin is associated with significant toxicity
Alternative therapy options
Susceptible Echinocandin Monotherapy Candidemia Clinically Stable, Clinically Unstable Option for isolates with known variable echinocandin susceptibility in patients who cannot tolerate amphotericin products. Combination of a susceptible echinocandin with a synergistic azole may be considered.
Echinocandin switch may be hindered by single echinocandin formularies.
Azole Monotherapy All sites Clinically Stable Option should be used as a step down agent when azole susceptibility is known. It is reasonable to start with an amphotericin product then de-escalate to an azole to complete the duration of treatment after the patient is clinically improving.
Azole Resistant Aspergillus spp.*
First-line therapy
Lipid Formulation Amphotericin-B Monotherapy All sites Clinically Stable, Clinically Unstable Recommended first-line treatment for azole-resistant A. fumigatus. Amphotericin products should serve as the backbone of therapeutic regimens and can be used in combination with other agents based on site of infection. Lipid Formulation Amphotericin-B is commonly recommended for IPA 3–5mg/kg/day. CNS aspergillosis is liposomal product at 5 mg/kg/day.
Amphotericin is associated with significant toxicity.
Alternative therapy options
Liposomal Amphotericin-B plus High-Dose Azole
Combination Regimen
All sites Clinically Stable, Clinically Unstable High-dose posaconazole and isavuconazole are potential additions to amphotericin products for combination therapy. Data suggests viability of high-dose azole monotherapy in select populations, but currently should be avoided in clinically unstable patients.
Azole plus Echinocandin
Combination Regimen
IPA Clinically Stable Voriconazole (standard dose) and an echinocandin is recommended as an empiric option in geographic regions with known resistance over 5%.
If used in patients with known azole-resistant A. fumigatus high-dose posaconazole or high-dose isavuconazole should be considered.
Echinocandin
Monotherapy
IPA Not recommended Echinocandin monotherapy is not recommended based on failure rates of 50% in non-azole resistant A. fumigatus.
Liposomal Amphotericin-B plus
High-Dose Azole
Or
Flucytosine
Or
Echinocandin
Combination Regimen
CNS Clinically Stable, Clinically Unstable In CNS aspergillosis, an amphotericin product should serve as the core agent for treatment. Experts recommend adding flucytosine, a high-dose azole, or an echinocandin as adjunctive agents. Flucytosine requires an acidic pH for activity against Aspergillus spp. It is unlikely that voriconazole can be dosed to overcome MICs in azole-resistant A. fumigatus therefore reasonable to utilize isavuconazole or posaconazole. Echinocandins do not have appreciable penetration into the CNS.
*

In all patients being treated with itraconazole, voriconazole, or posaconazole, therapeutic drug monitoring is recommended to optimize drug exposure and minimize side-effects. High-dose azole therapy may target trough concentrations above standard published recommendations (e.g. posaconazole).

Abbreviations: CNS, central nervous system; IPA, invasive pulmonary aspergillosis; TDM, therapeutic drug monitoring

Candidemia

Suspected or Known Resistance

When treating candidemia that is either suspected or known to have echinocandin resistance (prior echinocandin exposure, inability to clear blood cultures, clinically worsening), initiation of lipid formulation amphotericin-B (LF-AMB) at a dose of 3–5 mg/kg daily is recommended [7]. LF-AMB is currently a mainstay initial option for ERC since it binds ergosterol, is rapidly fungicidal, and is effective for invasive Candida infections. AMB products remain the only monotherapy option for ERC resistant to all echinocandins and azoles (Table 2). Liposomal amphotericin (LAMB) has been shown to have similar effects as compared to echinocandins on the reduction of cell viability in the biofilms and in antibiotic lock therapies of C. albicans strains [24]. A case series described poor outcomes in patients with ERC candidemia with mixed azole susceptibility patterns and full susceptibility to AMB. Authors concluded that delay in identification of resistance led to a delay in appropriate therapy contributing to death [25]. Although LF-AMB is recommended as the backbone of therapy for confirmed ERC, it has an extensive toxicity profile and may be intolerable for many patients. Therefore, alternative monotherapy or combination therapies may be considered as options in both early and late treatment of ERC.

Mixed Echinocandin Susceptibility

Some isolates of Candida may be able to be treated with an echinocandin depending on which specific mutations are expressed since not all echinocandin resistance displays a class effect [8, 10•]. It is reasonable to switch echinocandin monotherapy in patients with a known susceptibility to another echinocandin, especially with concomitant azole resistance when LF-AMB cannot be tolerated. However, it is important to note that many isolates with FKS mutations are resistant to two or more echinocandins [10•].

Azole Susceptible

Not all ERC strains are also resistant to azoles, leaving azoles as an option for candidemia if the isolate is susceptible. Using fluconazole in patients who have ERC poses some concern since it is suspected that many isolates may be intrinsically resistant (C. krusei), display lesser susceptibility (C. glabrata), or aforementioned fluconazole-echinocandin co-resistance. Voriconazole, posaconazole, and isavuconazole may retain activity against many ERC isolates but susceptibility should be confirmed prior to step-down therapy, especially for C. glabrata. Voriconazole has been compared to AMB for 3 to 7 days followed by fluconazole and has been shown to be equally as effective in clearing bloodstream infections in non-ERC isolates [26]. Although susceptibility to voriconazole was not noted, successful use of voriconazole was reported in a retrospective analysis of echinocandin-resistant C. glabrata infections over a 10-year period [27].

Combination Therapies

In vitro activity of echinocandins combined with other antifungals against echinocandin-resistant and fluconazole-echinocandin-resistant isolates of C. glabrata has been tested via checkerboard method to evaluate the fractional inhibitory concentration index of antifungal combinations. Synergism was noted with the combination of caspofungin plus posaconazole, anidulafungin plus posaconazole, and anidulafungin plus voriconazole in 85%, 70%, and 70% of the multidrug-resistant isolates, respectively. Though all studied isolates were sensitive to AMB by MIC, a lack of synergy was noted with the combination of an echinocandin plus AMB [28••]. Although these results are promising for synergistic effects and provide a future direction for research, these combinations have not been well studied in humans. Though we caution a broad application to clinical use, the aforementioned echinocandin-azole combinations may provide a therapeutic option when LF-AMB cannot be used. Successful use of posaconazole plus LF-AMB has also been noted in a retrospective analysis of C. glabrata infections over a period of 10 years [27].

Other Sites of Infection

Guidelines recommend treating patients with native and prosthetic valve Candida endocarditis or implantable cardiac device candidiasis with LF-AMB with or without 5-FC as an initial therapy option, which remains appropriate for ERC. This regimen may be eventually stepped down to an azole in clinically stable patients with azole-susceptible isolates that have cleared blood cultures. Utilizing an azole, when appropriate criteria are met, allows for an oral option to prolong therapy or provide suppressive therapy as may be required in cases of prosthetic valve Candida endocarditis [7]. Although this step-down approach may remain viable when azole susceptibility is confirmed in ERC, specific consideration to the azole choice and Candida spp. is required. Differential susceptibility can occur in biofilms as highlighted by a case describing a poor outcome of a patient with ERC endocarditis. ERC was not confirmed until after failing micafungin plus fluconazole and surgical resection allowed for a direct valve culture. This case highlights the potential for ERC subtypes to exist in biofilms, the importance of isolate susceptibility testing and source control [29].

In patients with ERC osteomyelitis, septic arthritis, and suppurative thrombophlebitis, source control should be evaluated and therapy with fluconazole or an alternative azole considered (if susceptible). LF-AMB remains a viable option for ERC and may be preferred as initial therapy. Step-down or suppressive therapy with an azole should be considered after appropriate response and proven susceptibility [7]. Case reports have described ERC septic arthritis and osteomyelitis successfully treated with LF-AMB for 4 weeks followed by voriconazole for 6 months [30]. Based on this case report and guideline recommendations, it is reasonable to do an extended course of LF-AMB followed by a long suppressive course of voriconazole for treatment of septic arthritis and osteomyelitis caused by ERC susceptible to voriconazole.

Candida auris

Especially concerning is the emergence of C. auris, a rare but commonly fluconazole resistant, frequently itraconazole, voriconazole, isavuconazole resistant, and variable AMB susceptible pathogen. Echinocandins remain the empiric treatment of choice; however, reports of echinocandin resistance and non-responsiveness are rising. Furthermore, the site of infection may reduce the activity of an echinocandin. Reports of persistent echinocandin-resistant C. auris urinary tract infections are emerging [11, 14]. Although it is rare for urinary tract infections to convert to candidemia, treatment options are limited if this were to occur. Echinocandins do not penetrate the urine well and thus 5-FC has been recommended as combination therapy for C. auris urinary tract infections due to its high bladder penetration [31]. AMB plus 5- FC has also been suggested but no clinical outcomes were reported [32]. Combination therapy such as conventional AMB with or without 5-FC may be used in urinary tract azole-resistant ERC infections that require antifungal therapy [7, 14, 32].

Echinocandin-resistant C. auris leaves the clinician with minimal options. LF-AMB remains the alternative therapy although 30% of isolates in the United States may be resistant [31, 33]. 5-FC may have variable activity and should be used in combination therapy only. Other reported combinations, such as micafungin plus voriconazole, may provide synergy though there is minimal clinical reports of successfully using this combination in echinocandin-resistant C. auris [34]. Pan-resistant C. auris has been reported following treatment [31]. In addition to aggressive source control for echinocandin- resistant C. auris, LF-AMB or a combination of active antifungals (micafungin plus voriconazole) may be reasonable.

Azole-Resistant Aspergillosis

Background/Epidemiology

Aspergillus spp. are the most common cause of invasive mold infections worldwide [35•]. Cases of azole-resistant Aspergillus fumigatus (ARAF) have been reported since the 1990s and have steadily increased over the past two decades [36]. Large epidemiologic studies report overall ARAF rates ranging from 3.2 to 27.6% of isolates, with higher resistance rates in Europe [37, 38]. The United States remains largely unaffected by resistance [39], but clinical occurrences have been documented since the mid-2000s [40]. A recent comprehensive review of ARAF epidemiology has been published elsewhere [35•, 40].

Two primary risks of developing ARAF include environmental exposure to azole-based fungicides and long-term use of azole antifungal therapy [40, 41]. Azole fungicides are commonly used in agriculture internationally and may pose a public health treat by introducing resistance mechanisms in Aspergillus spp. [42]. Previous studies have reported that 64% of patients with invasive ARAF never received an azole, suggesting an important role of environmental exposures [42, 43]. Additionally, clinical use of long-term or suppressive azole therapy exerts pressure on the organism which may lead to the development of resistance within a given patient. Azoles (voriconazole and isavuconazole) are a preferred mainstay of therapy for most invasive aspergillosis due to proven efficacy, tolerability, and because they offer an option for oral therapy [44, 45]. Therefore, the management of ARAF is especially concerning and requires thoughtful regimen selection based on patient evaluation.

Mechanism of Resistance

Triazoles work by inhibiting the fungal cytochrome lanosterol 14-α-demethylase, which is encoded by the CYP51A gene. 14-α-demethylase inhibition leads to an accumulation of 14-α-methyl sterols, a lack of functional ergosterol, and subsequently destabilizes the fungal cell membrane. The most common mechanisms of ARAF are mediated by mutations in the CYP51A gene (Table 1).

Oftentimes, mutations in the CYP51A gene are seen in combination. The most common mutation combination is the presence of a 34-base pair tandem repeat (TR34) in the promoter region of the CYP51A gene, which is seen with a substitution of leucine 98 to histidine (L98H), denoted as TR34/L98H. The TR34/L98H mutation leads to an overexpression of CYP51A [35•, 46]. Another frequent mutation combination is a 46-base pair tandem repeat in the CYP51A promoter region (TR46) with a substitution at tyrosine 121 for phenylalanine (Y121) and threonine 289 for alanine (T289A), referred to as TR46/Y121/ T289A [36]. The TR34/L98H and TR46/Y121/T289A mutations confer pan-azole resistance and yield difficult to treat infections. Point mutations, like substitutions at M220 and G448S, also lead to resistance and can propagate resistance to one or more azole antifungal [47]. For example, the G54 and G138 point mutation confer resistance primarily to itraconazole and posaconazole, while point mutations at G448 lead to voriconazole resistance, with possible resistance to itraconazole and posaconazole [35•, 48, 49, 50] (Table 1).

Non-CYP-mediated mechanisms of resistance include two efflux pumps, ATP-binding cassette (ABC) and major facilitator superfamily (MFS) transporters, and hmg1 and erg6 enzyme dysregulation [46, 51]. Mechanistically, overexpression of transmembrane pumps may not allow for appropriate intracellular drug concentrations to be obtained [52]. Comparatively, mutations in hmg1 and erg6 enzymes increase ergosterol synthesis and ultimately lead to a need for an increased amount of azole in order to inhibit ergosterol synthesis [51] (Table 1).

Identification

One barrier to successful treatment of ARAF is the irregularity of positive culture data for many clinical infections. ARAF is often identified late in the treatment course, if at all, and amplifies a poor prognosis as mortality rates of 88% in patients are documented [35•]. Means of identification are limited to EUCAST and CLSI microdilution, azole-based agar plates, and commercially available (but not clinically validated) polymerase chain reaction (PCR) tests. Some experts recommend following biomarkers (e.g., galactomannan) to infer persistence of infection in patients with documented aspergillosis [40], or assessing isolates for resistance and drug susceptibility in areas of known resistance [4••]. In most situations, the above information will not be immediately available to the clinician. This leads the clinician to work off of assumptions of geographic resistance patterns, prior triazole exposure, or persistence and worsening of infection despite optimized therapy.

Treatment

Invasive Pulmonary Aspergillosis

Empiric Treatment

The treatment of invasive pulmonary aspergillosis (IPA) due to ARAF is complicated and requires host-specific considerations such as immunosuppression, drug tolerability, and severity of infection [53, 54, 55]. In 2015, an expert consensus statement was published on ARAF management [4••]. Opinion-based recommendations were made for initial therapy based on known regional resistance rates to azoles considering the site of infection. Initiation of guideline-directed therapy, specifically voriconazole for IPA, in regions with documented ARAF rates less than 5% was recommended. In regions with ARAF rates of 5–10%, expert opinion suggested initial regimens of voriconazole alone, voriconazole plus an echinocandin, or LAMB alone. In contrast, LAMB or the combination of an echinocandin plus voriconazole should be initiated in areas of ARAF > 10%. These opinions reflect a pragmatic approach weighing the risk of resistance with the risk of toxicities associated with LAMB. Though we agree with this approach, we suggest isavuconazole as a reasonable alternative where voriconazole would otherwise be utilized.

Known Resistance or Failing Current Azole Therapy

Both American and European practice guidelines for the treatment of aspergillosis recommend voriconazole (or isavuconazole) as first-line therapy in patients with IPA [44, 45]. In cases of suspected or known ARAF, clinicians should consider a stepwise approach to treatment based on the clinical scenario. This includes source control, and optimizing current azole therapy (if ARAF not confirmed), switching treatment to a LF-AMB product, or exploring combination regimens (Table 2). In all cases, choosing an antifungal (alone or in combination) that ARAF is susceptible to is recommended by the aforementioned practice guidelines.

Dose Optimization of Azole Therapy

Therapeutic Drug Monitoring

Many azoles have known pharmacokinetic variability and are prone to drug interactions. Optimizing therapy through therapeutic drug monitoring (TDM), agent selection with consideration to the site of infection, and evaluating patient-specific administration factors should be assessed in all patients. Appropriate treatment trough concentration targets for azoles may vary based on the infection but are generally considered: itraconazole 1–4 mg/L, voriconazole 2–5.5 mg/L, and posaconazole 1–3.75 mg/L [45, 56]. Trough concentrations should be drawn after an appropriate time period for accuracy: voriconazole trough levels after 2–5 days and itraconazole and posaconazole trough levels after 5–7 days of therapy. Although not currently utilized in clinical practice, isavuconazole may have assigned TDM in the future [45, 57, 58]. In otherwise stable patients where ARAF is not confirmed but response to therapy is slow, optimizing azole therapy should be considered prior to switching therapy. We recommend TDM to optimize therapy in all patients to ensure subclinical drug exposure does not lead to treatment failure.

High-Dose Posaconazole or Isavuconazole Therapy

Although not recommended in the expert consensus statement, dose escalation of posaconazole or isavuconazole, based on MIC, may be a treatment option. Investigators have evaluated MIC distribution of clinical A. fumigatus isolates to determine susceptibility profiles of wild-type (WT), T34/L98H and TR46/ Y121F/T289A CYP51A mutations [59]. Of 363 isolates, 141 presented as non-WT. Of those non-WT isolates, the majority (88/141) had the T34/L98H mutation. Notably, 10% of the non-WT isolates presented with no CYP51A mutation. The MIC distributions varied based on type of mutation. The T34/L98H mutation conferred the following MICs when tested by CLSI and EUCAST methods: itraconazole > 8 mg/L, voriconazole 2–16 mg/L, and posaconazole 0.25–2 mg/L. The TR46/Y121F/T289A mutation had the following MIC distribution: itraconazole > 16 mg/L, voriconazole > 8 mg/L, and posaconazole 0.25–4 mg/L. The CLSI epidemiologic cut-off values are 1 mg/L for itraconazole and voriconazole, and 0.25 mg/L for posaconazole. The lesser MIC variability and distribution observed for posaconazole suggests it could potentially be used to treat ARAF at high doses (and TDM targets). Some in vivo models have shown promise with this practice [60]. Furthermore, a case series of 7 patients with ARAF intentionally treated with high-dose posaconazole targeting trough concentrations >3 mg/L reported survival of 4 patients and 3 that died from their underlying disease [61••].

An in vitro study assessed high-dose isavuconazole in ARAF isolates using the EUCAST clinical breakpoints [62, 63, 64]. Using a Monte Carlo simulation, investigators found doubling the dose of isavuconazole (400 mg of active drug three times daily, followed by 400 mg once daily [isavuconazonium 744 mg = 400 mg isavuconazole]) had a 90% chance of achieving target attainment of the assigned epidemiologic cut-off of 2 mg/L [62, 63]. The EUCAST clinical breakpoints published for Aspergillus spp. define susceptibility for isavuconazole ≤ 1 mg/L [64].

Clinicians must understand that the use of higher azole exposure to overcome MICs is an interesting option and may be reasonable in some situations but must also consider that not all laboratory assumptions fully or successfully translate into clinical practice [65]. Though reports show promise, careful consideration of patient response, infection site, and pathogen/antifungal MIC is required before utilizing this approach as a step-down option. At this time, high-dose azole therapy, if used, should be used in combination with another antifungal, especially in patients who are critically ill.

Pharmacotherapy Changes—Monotherapy

In patients with ARAF who are unstable/critically ill, LF-AMB remains a recommended therapy (Table 2) [4••, 45]. An experimental murine model of ARAF concluded that LAMB retains activity in the presence of azole resistance mechanisms [4••, 66]. These investigators also assessed the impact of host factors on the efficacy of LAMB in a murine model [67]. Regardless of immunosuppression status, LAMB remained efficacious in the treatment of ARAF but they observed that higher daily doses of LAMB may be required. The practical implications of these studies support the use of LAMB in the treatment of ARAF though higher dosing/exposure has not been clinically validated. The timely switch to LF-AMB in cases of suspected or known ARAF is warranted in unstable/critically ill patients. Special consideration should be made in cases of azole-resistant A. terreus and other species intrinsically resistant to AMB, especially as documented pathogenic clinical isolates are beginning to emerge [68].

Consideration may be given to echinocandin monotherapy, specifically caspofungin, as activity may be retained. However, documented failure rates of 50% or higher in azole-susceptible A. fumigatus is unacceptably high [69, 70, 71]. We recommend combination antifungal therapy if an echinocandin is used. Overall, there is a paucity of clinical evidence and more data is needed to support the appropriate combination treatments for ARAF.

Pharmacotherapy Changes—Combination Therapies

A panel of experts recommend the combination of voriconazole and an echinocandin as an option for ARAF [4••]. This recommendation is based on preclinical data that demonstrated increased susceptibility in ARAF strains [72, 73]. The use of combination therapy with voriconazole and anidulafungin has shown a trend towards mortality benefit in patients with hematologic malignancies or hematopoietic stem cell transplant with invasive Aspergillus [74]. However, ARAF was not specifically addressed.

Since the publication of the expert consensus statement, in vitro data has questioned echinocandin-azole combination therapy. Isavuconazole in combination with anidulafungin, caspofungin, or micafungin in 30 Aspergillus strains, including 5 ARAF isolates, ultimately showed no synergism (5/5 indifference) with combination for ARAF isolates [75]. An ongoing clinical trial evaluating the use of a PCR assay for identifying resistance mechanisms in invasive aspergillosis is advising a therapeutic change to LAMB 3 mg/kg or the combination of an echinocandin plus posaconazole, with higher goal posaconazole trough concentrations of 3–4 mg/L (see “High Dose Posaconazole or Isavuconazole Therapy” above) when toxicity precludes the use of LAMB, if resistance is detected [76]. This study may provide much needed data on the use of combination and higher trough-driven posaconazole therapy for the management of ARAF.

Central Nervous System Infections

Central nervous system (CNS) aspergillosis has a dismal prognosis and very limited efficacy data to draw a best treatment approach for ARAF [77]. Current guidelines recommend voriconazole as the treatment of choice in susceptible isolates [44]. LAMB at a dose of 5 mg/kg/day is the mainstay of therapy for CNS ARAF due to its ability to penetrate brain parenchymal tissue. All experts in the consensus statement agreed that a second agent should be added, including 5-FC, voriconazole, or an echinocandin. While credence was given specifically to 5-FC because of adequate CSF penetration, it has minimal A. fumigatus activity at a pH of 7 [4••, 78]. Optimal voriconazole dosing for CNS ARAF is unknown, but one anticipates dose-limiting toxicities with higher drug exposures even with close TDM. Other agents, like posaconazole or isavuconazole, have limited clinical data to support their use in CNS aspergillosis [79, 80, 81]. Although cerebral spinal fluid concentrations are low for these agents, parenchymal tissue concentrations are noted to be adequate [73, 75, 82]. Echinocandins are an adjunct treatment option, but these agents have poor CNS penetration and are not recommended as monotherapy [4••].

Other Sites of Infection

Other types of infection, like endophthalmitis, endocarditis, osteomyelitis, peritonitis, or pyelonephritis/cystitis, have minimal specific clinical data to update in terms of ARAF. It can be inferred from the aforementioned discussion that published expert consensus recommendations can be followed for these sites of infection in regard to dose and formulation of AMB product or adjunctive agents as reviewed elsewhere [44].

Expert Opinion

Although positive clinical outcomes with the treatment regimens outlined above have been noted in literature, robust prospective studies are needed to determine the optimal therapy for treatment of ERC and ARAF. Delaying appropriate therapy can lead to patient mortality and morbidity; therefore, prompt recognition and optimization are critical.

Currently, LF-AMB serves as a backbone initial therapy for the treatment of known or suspected ERC, especially in clinically unstable patients. Other treatment options are outlined in Table 2. Although combination therapy for ERC is promising, data is limited outside of specific clinical scenarios.

ARAF has clear documentation of high mortality rates and is rarely recognized early in the treatment course. In patients with known ARAF that are unstable or acutely ill, LF-AMB is the preferred therapy. Frequently, treatment-limiting toxicities and sub-optimal response with LF-AMB necessitate exploring alternative and combination therapies, respectively, as outlined in Table 2. Patient-specific factors, source control, and azole-specific MICs should be considered prior to therapy modification. An ongoing clinical trial may help with unanswered questions on optimal treatment approaches, including step-down therapy options [76].

Conclusion

Fungal resistance is an emerging health crisis requiring prompt evaluation and advanced knowledge of antifungal pharmacology to optimally manage infections. Current treatment options are limited and rely on LAMB for both ERC and ARAF. As the newer therapies emerge, treatment recommendations for ERC and ARAF are likely to shift based on safety and efficacy profile of the new agents. Future research defining optimal therapies and early identification of ERC and ARAF is of extreme importance.

Acknowledgments

Funding: Study authors are supported by the Colorado Clinical and Translational Sciences Institute (CCTSI). The CCTSI is supported in part by Colorado CTSA Grant UL1TR002535 from NCATS/NIH.

Tyree Kiser reports grants from NCATS/NIH CTSA Grant UL1TR002535 during the conduct of the study; and grants from Astellas, grants from Allergan and grants from Pfizer outside the submitted work. Scott Mueller reports grants from NCATS/NIH CTSA Grant UL1TR002535, which supports the Colorado Clinical and Translational Sciences Institute; from CSL Behring Investigator-Initiated Study, grants from Merck Investigator-Initiated Studies Program; all industry funding has been received by the institution that employees Dr. Mueller, and is outside the submitted work (unrelated to antifungal therapeutics). Alison Novak and Mary Bradley declare no conflicts of interest relevant to this manuscript.

Footnotes

Compliance with Ethics Guidelines

Conflict of Interest

Disclosures: Dr. Kiser discloses investigator initiated research funding from Astellas, Pfizer, and Allergan. Dr. Mueller discloses investigator initiated research funding from CSL Behring and Merck. All other authors have no relevant disclosures.

Human and Animal Rights and Informed Consent

This article does not contain any studies with human or animal subjects performed by any of the authors.

References

Papers of particular interest, published recently, have been highlighted as:

• Of importance

•• Of major importance

  • 1.Sanguinetti M, Posteraro B, Lass-Flörl C. Antifungal drug resis- tance among Candida species: mechanisms and clinical impact. Mycoses. 2015;58:2–13. 10.1111/myc.12330. [DOI] [PubMed] [Google Scholar]
  • 2.Perlin DS. Echinocandin resistance in Candida. Clin Infect Dis.2015;61:S612–7. 10.1093/cid/civ791. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Matzaraki V, Gresnigt MS, Jaeger M, Ricaño-Ponce I, Johnson MD, Oosting M, et al. An integrative genomics approach identifies novel pathways that influence candidaemia susceptibility. PLoS One. 2017;12:e0180824 10.1371/journal.pone.0180824. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Verweij PE, Ananda-Rajah M, Andes D, Arendrup MC, Brüggemann RJ, Chowdhary A, et al. International expert opinion on the management of infection caused by azole-resistant Aspergillus fumigatus. Drug Resist Updat. 2015;21–22:30–40. https://linkinghub.elsevier.com/retrieve/pii/S1368764615000357. Accessed October 30, 2019.••Expert consensus statement focused specifically on the empiric and definitive treatment of azole- resistant Aspergillus fumigatus.
  • 5.Wiederhold N. Antifungal resistance: current trends and future strategies to combat. Infect Drug Resist. 2017;10:249–59. 10.1016/j.drup.2015.08.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Pais P, Galocha M, Viana R, Cavalheiro M, Pereira D, Teixeira MC. Microevolution of the pathogenic yeasts Candida albicans and Candida glabrata during antifungal therapy and host infection. Microb Cell. 2019;6:142–59. 10.15698/mic2019.03.670. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Pappas PG, Kauffman CA, Andes DR, Clancy CJ, Marr KA, Ostrosky-Zeichner L, et al. Clinical practice guideline for the man- agement of candidiasis: 2016 update by the Infectious Diseases Society of America. Clin Infect Dis. 2015;civ933 10.1093/cid/civ933 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Arendrup MC, Patterson TF. Multidrug-resistant candida: epidemiology, molecular mechanisms, and treatment. J Infect Dis.2017;216:S445–51. 10.1093/infdis/jix131. [DOI] [PubMed] [Google Scholar]
  • 9.Vallabhaneni S, Cleveland AA, Farley MM, Harrison LH, Schaffner W, Beldavs ZG, et al. Epidemiology and risk factors for echinocandin nonsusceptible Candida glabrata bloodstream infections: Data from a large multisite population-based candidemia surveillance program, 2008–2014. Open Forum Infect Dis. 2015;2(4):ofv163 10.1093/ofid/ofv163 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Pfaller MA, Diekema DJ, Turnidge JD, Castanheira M, Jones RN. Twenty years of the sentry antifungal surveillance program: results for Candida species from 1997–2016. Open Forum Infect Dis. 2019;6:S79–94. 10.1093/ofid/ofy358.•Recent comprehensive review for Candida spp. resistance patterns over a period of 20 years.
  • 11.Centers for Disease Control and Prevention. Recommendation for identification of Candida auris. https://www.cdc.gov/fungal/candida-auris/index.html. Accessed November 1, 2019.
  • 12.Public Health England. Guidance for the laboratory investigation, management and infection prevention and control for cases of Candida auris. https://assets.publishing.service.gov.uk/government/uploads/system/uploads/attachment_data/file/637685/Updated_Candida_auris_Guidance_v2.pdf. Accessed November 1, 2019.
  • 13.Hou X, Lee A, Jiménez-Ortigosa C, Kordalewska M, Perlin DS, Zhao Y. Rapid detection of erg11 -associated azole resistance and fks-associated echinocandin resistance in Candida auris. Antimicrob Agents Chemother. 2018;63:e01811–8. 10.1128/AAC.01811-18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Biagi MJ, Wiederhold NP, Gibas C, Wickes BL, Lozano V, Bleasdale SC, et al. Development of high-level echinocandin resistance in a patient with recurrent Candida auris candidemia secondary to chronic candiduria. Open Forum Infect Dis. 2019;6:ofz262 10.1093/ofid/ofz262/5510065 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Arendrup MC, Prakash A, Meletiadis J, Sharma C, Chowdhary A. Comparison of EUCAST and CLSI reference microdilution mics of eight antifungal compounds for candida auris and associated tentative epidemiological cutoff values. Antimicrob Agents Chemother. 2017;61:e00485–17. 10.1128/AAC.00485-17.••Recent publication of Candida auris MICs to interpret the extent of resistance although no clinical breakpoints have been set.
  • 16.Rivero-Menendez O, Navarro-Rodriguez P, Bernal-Martinez L, Martin-Cano G, Lopez-Perez L, Sanchez-Romero I, et al. Clinical and laboratory development of echinocandin resistance in Candida glabrata: Molecular characterization. Front Microbiol. 2019;10:1585 10.3389/fmicb.2019.01585. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Pham CD, Iqbal N, Bolden CB, Kuykendall RJ, Harrison LH, Farley MM, et al. Role of FKS mutations in Candida glabrata: mic values, echinocandin resistance, and multidrug resistance. Antimicrob Agents Chemother. 2014;58:4690–6. 10.1128/AAC.03255-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Bordallo-Cardona MÁ, Escribano P, de la Pedrosa GGE, Marcos- Zambrano LJ, Cantón R, Bouza E, et al. In vitro exposure to increasing micafungin concentrations easily promotes echinocandin resistance in Candida glabrata isolates. Antimicrob Agents Chemother. 2016;61(2):e01542–16. 10.1128/AAC.01542-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Beardsley J, Halliday CL, Chen SC-A, Sorrell TC. Responding to the emergence of antifungal drug resistance: Perspectives from the bench and the bedside. Future Microbiol. 2018;13:1175–91. 10.2217/fmb-2018-0059. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Jensen RH, Astvad KMT, Silva LV, Sanglard D, Jørgensen R, Nielsen KF, et al. Stepwise emergence of azole, echinocandin and amphotericin B multidrug resistance in vivo in Candida albicans orchestrated by multiple genetic alterations. J Antimicrob Chemother. 2015;70:2551–5. 10.1093/jac/dkv140. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Staab JF, Neofytos D, Rhee P, Jiménez-Ortigosa C, Zhang SX, Perlin DS, et al. Target enzyme mutations confer differential echinocandin susceptibilities in Candida kefyr. Antimicrob Agents Chemother. 2014;58:5421–7. 10.1128/AAC.00096-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Healey KR, Jimenez Ortigosa C, Shor E, Perlin DS. Genetic drivers of multidrug resistance in Candida glabrata. Front Microbiol. 2016;7 10.3389/fmicb.2016.01995. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Healey K, Perlin DS. Fungal resistance to echinocandins and the MDR phenomenon in Candida glabrata. J Fungi. 2018;4(3). 10.3390/jof4030105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Basas J, Palau M, Gomis X, Almirante B, Gavaldà J. Efficacy of liposomal amphotericin B and anidulafungin using an antifungal lock technique (ALT) for catheter-related Candida albicans and Candida glabrata infections in an experimental model. Coste AT, editor. PLoS One. 2019;14(2):e0212426 10.1371/journal.pone.0212426 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Chew KL, Octavia S, Lin RTP, Yan GZ, Teo JWP. Delay in effective therapy in anidulafungin-resistant Candida tropicalis fungaemia: Potential for rapid prediction of antifungal resistance with whole-genome-sequencing. J Glob Antimicrob Resist. 2019;16:105–7. 10.1016/j.jgar.2018.12.010. [DOI] [PubMed] [Google Scholar]
  • 26.Kullberg BJ, Sobel JD, Ruhnke M, Pappas PG, Viscoli C, Rex JH, et al. Voriconazole versus a regimen of amphotericin B followed by fluconazole for candidaemia in non-neutropenic patients: A randomised non-inferiority trial. Lancet. 2005;366(9495):1435–42. 10.1016/S0140-6736(05)67490-9. [DOI] [PubMed] [Google Scholar]
  • 27.Alexander BD, Johnson MD, Pfeiffer CD, Jiménez-Ortigosa C, Catania J, Booker R, et al. Increasing echinocandin resistance in Candida glabrata: Clinical failure correlates with presence of FKS mutations and elevated minimum inhibitory concentrations. Clin Infect Dis. 2013;56:1724–32. 10.1093/cid/cit136. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Denardi LB, Keller JT, Oliveira V, DAN M, Santurio JM, Alves SH. Activity of combined antifungal agents against multidrug-re- sistant Candida glabrata strains. Mycopathologia. 2017;182:819–28. 10.1007/s11046-017-0141-9.•• Recent checkerboard analysis of combined antifungal agents in vitro to predict the synergistic effect against C. glabrata.
  • 29.Agnelli C, Guinea J, Valerio M, Escribano P, Emilio B, Munoz P. Infectious endocarditis caused by Candida glabrata: Evidence of in vivo development of echinocandin resistance. Rev Esp Quimioter. 2019;34:395–7. [PMC free article] [PubMed] [Google Scholar]
  • 30.Wright WF, Bejou N, Shields RK, Marr K, McCarty TP, Pappas PG. Amphotericin B induction with voriconazole consolidation as salvage therapy for FKS - associated echinocandin resistance in Candida glabrata septic arthritis and osteomyelitis. Antimicrob Agents Chemother. 2019;63:e00512–9. 10.1128/AAC.00512-19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Chowdhary A, Sharma C, Meis JF. Candida auris: A rapidly emerging cause of hospital-acquired multidrug-resistant fungal infections globally. Hogan DA, editor. PLoS Pathog. 2017;13(5):e1006290 10.1371/journal.ppat.1006290 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Rhodes J, Abdolrasouli A, Farrer RA, Cuomo CA, Aanensen DM, Armstrong-James D, et al. Genomic epidemiology of the UK out- break of the emerging human fungal pathogen Candida auris. Emerg Microbes Infect. 2018;7(1):1–12. 10.1038/s41426-018-0045-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Centers for Disease Control and Prevention. Recommendation for identification of Candida auris. https://www.cdc.gov/fungal/candida-auris/c-auris-antifungal.html. Accessed November 14, 2019.
  • 34.Fakhim H, Chowdhary A, Prakash A, Vaezi A, Dannaoui E, Meis JF, et al. In vitro interactions of echinocandins with triazoles against multidrug-resistant Candida auris. Antimicrob Agents Chemother. 2017;61:e01056–17. 10.1128/AAC.01056-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Chowdhary A, Sharma C, Meis JF. Azole-resistant aspergillosis: epidemiology, molecular mechanisms, and treatment. J Infect Dis. 2017;216:S436–44. 10.1093/infdis/jix210.•Recent comprehensive epidemiology review for azole-resistant Aspergillus fumigatus.
  • 36.Denning DW, Venkateswarlu K, Oakley KL, Anderson MJ, Manning NJ, Stevens DA, et al. Itraconazole resistance in Aspergillus fumigatus. Antimicrob Agents Chemother. 1997;41:1364–8. 10.1128/AAC.41.6.1364. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Van der Linden JWM, Arendrup MC, Warris A, Lagrou K, Pelloux H, Hauser PM, et al. Prospective multicenter international surveil- lance of azole resistance in Aspergillus fumigatus. Emerg Infect Dis. 2015;21(6):1041–4. 10.3201/eid2106.140717. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Lockhart SR, Frade JP, Etienne KA, Pfaller MA, Diekema DJ, Balajee SA. Azole resistance in aspergillus fumigatus isolates from the artemis global surveillance study is primarily due to the TR/ l98H mutation in the cyp51A Gene. Antimicrob Agents Chemother. 2011;55:4465–8. 10.1128/AAC.00185-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Pham CD, Reiss E, Hagen F, Meis JF, Lockhart SR. Passive surveillance for azole-resistant Aspergillus fumigatus, United States, 2011–2013. Emerg Infect Dis. 2014;20(9):1498–503. 10.3201/eid2009.140142. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Verweij PE, Chowdhary A, Melchers WJG, Meis JF. Azole resistance in Aspergillus fumigatus : Can we retain the clinical use of mold-active antifungal azoles? Weinstein RA, editor. Clin Infect Dis. 2016;62:362–8. 10.1093/cid/civ885. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Camps SMT, van der Linden JWM, Li Y, Kuijper EJ, van Dissel JT, Verweij PE, et al. Rapid induction of multiple resistance mechanisms in aspergillus fumigatus during azole therapy: A case study and review of the literature. Antimicrob Agents Chemother. 2012;56:10–6. 10.1128/AAC.05088-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Snelders E, Camps SMT, Karawajczyk A, Schaftenaar G, Kema GHJ, van der Lee HA, et al. Triazole fungicides can induce cross- resistance to medical triazoles in Aspergillus fumigatus. Feldmesser M, editor. PLoS One. 2012;7(3):e31801 10.1371/journal.pone.0031801 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Van der Linden JWM, Snelders E, Kampinga GA, Rijnders BJA, Mattsson E, Debets-Ossenkopp YJ, et al. Clinical implications of azole resistance in Aspergillus fumigatus, the Netherlands, 2007–2009. Emerg Infect Dis. 2011;17(10):1846–54. 10.3201/eid1710.110226. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Patterson TF, Thompson GR, Denning DW, Fishman JA, Hadley S, Herbrecht R, et al. Practice guidelines for the diagnosis and management of Aspergillosis: 2016 Update by the Infectious Diseases Society of America. Clin Infect Dis. 2016;63:e1–60. 10.1093/cid/ciw326. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Ullmann AJ, Aguado JM, Arikan-Akdagli S, Denning DW, Groll AH, Lagrou K, et al. Diagnosis and management of Aspergillus diseases: Executive summary of the 2017 ESCMID-ECMM-ERS guideline. Clin Microbiol Infect. 2018;24:e1–38. 10.1016/j.cmi.2018.01.002. [DOI] [PubMed] [Google Scholar]
  • 46.Pérez-Cantero A, López-Fernández L, Guarro-Artigas J, Capilla J.Azole resistance mechanisms in Aspergillus: Update and recent advances. Int J Antimicrob Agents. 2019;105807 10.1016/j.ijantimicag.2019.09.011. [DOI] [PubMed] [Google Scholar]
  • 47.Arikan-Akdagli S, Ghannoum M, Meis J. Antifungal resistance: specific focus on multidrug resistance in Candida auris and secondary azole resistance in Aspergillus fumigatus. J Fungi. 2018;4(4):129 10.3390/jof4040129. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Diaz-Guerra TM, Mellado E, Cuenca-Estrella M, Rodriguez- Tudela JL. A point mutation in the 14-alpha-sterol demethylase gene cyp51A contributes to itraconazole resistance in Aspergillus fumigatus. Antimicrob Agents Chemother. 2003;47(3):1120–4. 10.1128/aac.47.3.1120-1124.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Nascimento AM, Goldman GH, Park S, Marras SAE, Delmas G, Oza U, et al. Multiple resistance mechanisms among Aspergillus fumigatus mutants with high-level resistance to itraconazole. Antimicrob Agents Chemother. 2003;47:1719–26. 10.1128/AAC.47.5.1719-1726.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Howard SJ, Webster I, Moore CB, Gardiner RE, Park S, Perlin DS, et al. Multiazole resistance in Aspergillus fumigatus. Int J Antimicrob Agents. 2006;28:450–3. 10.1016/j.jantimicag.2006.08.017. [DOI] [PubMed] [Google Scholar]
  • 51.Hagiwara D, Arai T, Takahashi H, Kusuya Y, Watanabe A, Kamei K. Azole-Resistant Aspergillus fumigatus Isolates with mutation in HMG-CoA Reductase. Emerg Infect Dis. 2018;24:1889–97. 10.3201/eid2410.180730. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Perlin DS, Shor E, Zhao Y. Update on antifungal drug resistance. Curr Clin Microbiol Rep. 2015;2:84–95. 10.1007/s40588-015-0015-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Bellanger A-P, Berceanu A, Scherer E, Desbrosses Y, Daguindau E, Rocchi S, et al. Invasive fungal disease, isavuconazole treatment failure, and death in acute myeloid leukemia patients. Emerg Infect Dis. 2019;25:1778–9. 10.3201/eid2509.190598. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Pilmis B, Garcia-Hermoso D, Alanio A, Catherinot E, Scemla A, Jullien V, et al. Failure of voriconazole therapy due to acquired azole resistance in Aspergillus fumigatus in a kidney transplant recipient with chronic necrotizing aspergillosis. Am J Transplant. 2018;18:2352–5. 10.1111/ajt.14940. [DOI] [PubMed] [Google Scholar]
  • 55.Van Ingen J, van der Lee HAL, Rijs AJMM, Snelders E, Melchers WJG, Verweij PE. High-level pan-azole-resistant Aspergillosis: TABLE 1. Warnock DW, editor. J Clin Microbiol. 2015;53:2343–5. 10.1128/JCM.00502-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Lewis RE. Current concepts in antifungal pharmacology. Mayo Clin Proc. 2011. [cited 2019 Nov 12];86:805–817. https://linkinghub.elsevier.com/retrieve/pii/S0025619611651835 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Andes D, Kovanda L, Desai A, Kitt T, Zhao M, Walsh TJ. Isavuconazole concentration in real-world practice: Consistency with results from clinical trials. Antimicrob Agents Chemother. 2018;62:e00585–18. 10.1128/AAC.00585-18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Stott KE, Hope WW. Therapeutic drug monitoring for invasive mould infections and disease: Pharmacokinetic and pharmacodynamic considerations. J Antimicrob Chemother. 2017;72(1):i12–8. 10.1093/jac/dkx029. [DOI] [PubMed] [Google Scholar]
  • 59.Buil J, Hagen F, Chowdhary A, Verweij P, Meis J. Itraconazole, voriconazole, and posaconazole CLSI MIC distributions for wild- type and azole-resistant Aspergillus fumigatus isolates. J Fungi. 2018;4:103 10.3390/jof4030103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Lepak AJ, Marchillo K, VanHecker J, Andes DR. Posaconazole pharmacodynamic target determination against wild-type and cyp51 mutant isolates of aspergillus fumigatus in an in vivo model of invasive pulmonary aspergillosis. Antimicrob Agents Chemother. 2013;57:579–85. 10.1128/AAC.01279-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Schauwvlieghe AFAD, Buil JB, Verweij PE, Hoek RAS, Cornelissen JJ, Blijlevens NMA, et al. High-dose posaconazole for azole-resistant aspergillosis and other difficult-to-treat mould infections. Mycoses. 2019;myc.13028 10.1111/myc.13028.•• Recent clinical case series on the success of using high-dose posaconazole therapy for treatment of invasive fungal infections, including azole-resistant Aspergillus fumigatus.
  • 62.Buil JB, Brüggemann RJM, Wasmann RE, Zoll J, Meis JF, Melchers WJG, et al. Isavuconazole susceptibility of clinical Aspergillus fumigatus isolates and feasibility of isavuconazole dose escalation to treat isolates with elevated MICs. J Antimicrob Chemother. 2018;73:134–42. 10.1093/jac/dkx354. [DOI] [PubMed] [Google Scholar]
  • 63.EUCAST. Isavuconazole and Aspergillus spp.; rationale for the clinical breakpoints. 2015;1–13. Retrieved from: http://www.eucast.org. Accessed November 12, 2019.
  • 64.EUCAST. Antifungal clinical breakpoint table. 2018. Retrieved from: http://www.eucast.org/fileadmin/src/media/PDFs/EUCAST_files/AFST/Clinical_breakpoints/Antifungal_breakpoints_v_9.0_180212.pdf. Accessed November 12, 2019.
  • 65.Stevens VM, Mueller SW, Reynolds PM, et al. Extrapolating antifungal animal data to humans—Is it reliable? Curr Fungal Infect Rep. 2020. 10.1007/s12281-020-00370-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Seyedmousavi S, Melchers WJG, Mouton JW, Verweij PE. Pharmacodynamics and dose-response relationships of liposomal amphotericin B against different azole-resistant Aspergillus fumigatus isolates in a murine model of disseminated aspergillosis. Antimicrob Agents Chemother. 2013;57:1866–71. 10.1128/AAC.02226-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Seyedmousavi S, Mouton JW, Melchers WJG, Verweij PE. In vivo efficacy of liposomal amphotericin B against wild-type and azole-resistant aspergillus fumigatus isolates in two different immunosuppression models of invasive aspergillosis. Antimicrob Agents Chemother. 2017;61:e02479–16. 10.1128/AAC.02479-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Zoran T, Sartori B, Sappl L, Aigner M, Sánchez-Reus F, Rezusta A, et al. Azole-resistance in aspergillus terreus and related species: an emerging problem or a rare phenomenon? Front Microbiol. 2018;9:516 10.3389/fmicb.2018.00516. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Viscoli C, Herbrecht R, Akan H, Baila L, Sonet A, Gallamini A, et al. An EORTC Phase II study of caspofungin as first-line therapy of invasive aspergillosis in haematological patients. J Antimicrob Chemother. 2009;64:1274–81. 10.1093/jac/dkp355. [DOI] [PubMed] [Google Scholar]
  • 70.Herbrecht R, Maertens J, Baila L, Aoun M, Heinz W, Martino R, et al. Caspofungin first-line therapy for invasive aspergillosis in allogeneic hematopoietic stem cell transplant patients: An European Organisation for Research and Treatment of Cancer study. Bone Marrow Transplant. 2010;45:1227–33. 10.1038/bmt.2009.334. [DOI] [PubMed] [Google Scholar]
  • 71.Cornely OA, Vehreschild JJ, Vehreschild MJGT, Würthwein G, Arenz D, Schwartz S, et al. Phase II dose escalation study of caspofungin for invasive aspergillosis. Antimicrob Agents Chemother. 2011;55:5798–803. 10.1128/AAC.05134-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Jeans AR, Howard SJ, Al-Nakeeb Z, Goodwin J, Gregson L, Warn PA, et al. Combination of voriconazole and anidulafungin for treatment of triazole-resistant aspergillus fumigatus in an in vitro model of invasive pulmonary aspergillosis. Antimicrob Agents Chemother. 2012;56:5180–5. 10.1128/AAC.01111-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Seyedmousavi S, Bruggemann RJM, Melchers WJG, Rijs AJMM, Verweij PE, Mouton JW. Efficacy and pharmacodynamics of voriconazole combined with anidulafungin in azole-resistant invasive aspergillosis. J Antimicrob Chemother. 2013;68:385–93. 10.1093/jac/dks402. [DOI] [PubMed] [Google Scholar]
  • 74.Marr KA, Schlamm HT, Herbrecht R, Rottinghaus ST, Bow EJ, Cornely OA, et al. Combination antifungal therapy for invasive aspergillosis: A randomized trial. Ann Intern Med. 2015;162:81 10.7326/M13-2508. [DOI] [PubMed] [Google Scholar]
  • 75.Raffetin A, Courbin V, Jullien V, Dannaoui E. In vitro combination of isavuconazole with echinocandins against azole-susceptible and -resistant aspergillus spp. Antimicrob Agents Chemother. 2017;62:e01382–17. 10.1128/AAC.01382-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Rijnders B PCR Based detection of azole resistance in A. fumigatus to improve patient outcome (AzorMan). https://clinicaltrials.gov/ct2/show/NCT03121235. [Google Scholar]
  • 77.Van der Linden JWM, Jansen RR, Bresters D, Visser CE, Geerlings SE, Kuijper EJ, et al. Azole-resistant central nervous system asper- gillosis. Clin Infect Dis. 2009;48:1111–3. 10.1086/597465. [DOI] [PubMed] [Google Scholar]
  • 78.Verweij P, Dorsthorst TE, Janssen DT, Meis WH, Mouton JW. In vitro activities at pH 5.0 and pH 7.0 and in vivo efficacy of flucytosine against Aspergillus fumigatus. Antimicrob Agents Chemother. 2008;52:4483–5. 10.1128/AAC.00491-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Schmitt-Hoffmann A-H, Kato K, Townsend R, Potchoiba MJ, Hope WW, Andes D, et al. Tissue distribution and elimination of isavuconazole following single and repeat oral-dose administration of isavuconazonium sulfate to rats. Antimicrob Agents Chemother. 2017;61:e01292–17. 10.1128/AAC.01292-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Lamoth F, Mercier T, André P, Pagani JL, Pantet O, Maduri R, et al. Isavuconazole brain penetration in cerebral aspergillosis. J Antimicrob Chemother. 2019;74(6):1751–3. 10.1093/jac/dkz050. [DOI] [PubMed] [Google Scholar]
  • 81.Nau R, Sorgel F, Eiffert H. Penetration of drugs through the blood- cerebrospinal fluid/blood-brain barrier for treatment of central nervous system infections. Clin Microbiol Rev. 2010;23:858–83. 10.1128/CMR.00007-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Rouzaud C, Jullien V, Herbrecht A, Palmier B, Lapusan S, Morgand M, et al. Isavuconazole diffusion in infected human brain. Antimicrob Agents Chemother. 2019;63:e02474–18. 10.1128/AAC.02474-18. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES