
Antifungal resistance: Emerging mechanisms and implications (Review)
- Authors:
- Published online on: July 8, 2025 https://doi.org/10.3892/mmr.2025.13612
- Article Number: 247
-
Copyright: © Kadariswantiningsih et al. This is an open access article distributed under the terms of Creative Commons Attribution License.
Abstract
Introduction
Antifungal resistance is an increasingly prominent issue in clinical medicine, primarily because of the rising incidence of fungal infection and limited arsenal of effective antifungal drugs. For example, the estimated global burden of invasive aspergillosis has risen from 250,000 in 2012 to 2.1 million in 2021, with only four classes of systemic antifungal are available-polyenes, azoles, echinocandins, and pyrimidine analogues (1–3). In 2020, diseases caused by fungi affected >1 billion individuals worldwide with 1.7 million death, comparable with the mortality caused by tuberculosis or human immunodeficiency virus (HIV) infection (1–4). Antifungal resistance is defined as the ability of fungal species to proliferate even in the presence of antifungal drug concentrations that would typically inhibit or kill the majority of isolates (4). Antifungal resistance leads to failure of treatment, prolonged infection and increased healthcare costs and morbidity and mortality rates (4). The prevalence of severe fungal infection has increased owing to the increasing population of immunocompromised individuals, such as those with diabetes, advanced cancer, HIV and old age (5).
Resistance in fungi can be intrinsic or acquired. Intrinsic resistance is a natural characteristic of certain fungal species, where antifungal agents are inherently ineffective due to pre-existing genetic traits (6) (for example, the intrinsic resistance of Cryptococcus species to echinocandins) (7). Species such as Aspergillus pluralis (spp)., Pichia kudriavzevii (formerly Candida krusei), Nakaseomyces glabratus (formerly C. glabrata) and certain strains of C. auris exhibit intrinsic resistance to fluconazole without prior exposure to the drug (8,9). By contrast, acquired resistance arises through genetic mutations and adaptations in response to antifungal exposure, allowing previously susceptible fungi to survive under drug pressure (2).
Antifungal tolerance further compounds the complexity of antifungal resistance. Antifungal tolerance involves a subpopulation of fungal cells that persist and proliferate slowly at drug concentrations higher than the minimum inhibitory concentration (MIC) (6). This tolerance can lead to treatment failure and is often mistakenly equated with resistance in the literature, although it represents a distinct adaptive strategy (10–12).
The present review summarizes current antifungal resistance and tolerance mechanisms, highlighting the genetic, physiological and epigenetic factors involved. Examining classes of mutations and their roles in the development of resistance may demonstrate how fungi adapt to antifungal pressures. Additionally, the present review aimed to explore the specific challenges and clinical implications of different types of resistance, particularly in the context of diagnosis and therapeutic limitations.
While previous reviews have addressed either genetic alterations, such as point mutations and aneuploidy (13–15), or epigenetic modulation through chromatin remodelling and histone modification (16,17), to the best of our knowledge, an integrated approach has not been performed. The present review aims to present a unified framework of both genetic and epigenetic drivers of antifungal resistance. Furthermore, the present review aimed to offer a translational perspective by linking these mechanistic insights with therapeutic strategies, including the use of histone deacetylase (HDAC) inhibitors, antifungal-loaded nanoparticles and stewardship approaches that aims to optimize treatment selection, dose and duration to minimize resistance. Understanding these mechanisms in an integrated manner is key for developing novel antifungal therapy and devising effective strategies to combat the increasing threat of drug-resistant fungal infection.
Intrinsic resistance
In fungi, intrinsic resistance refers to the innate ability of certain species to withstand the effects of specific antifungal agents without prior exposure or adaptation (18,19). This form of resistance is encoded within the genetic framework, making the species naturally less susceptible or entirely resistant to antifungal drugs (18,19). Several mechanisms underly intrinsic resistance (Table I).
Ineffective drug-target binding affinity
A key intrinsic resistance mechanism is the ineffective binding of antifungal drugs to targets. For example, variations in the structure of the enzyme sterol 14α-demethylases (SDMs) in certain fungi result in decreased binding affinity for azole antifungals, rendering these drugs ineffective (20). SDM is a key enzyme in fungal ergosterol biosynthesis. SDM catalyses the conversion of lanosterol or eburicol to a 14α-demethylated product (20). The binding of azole antifungals to SDM depletes ergosterol and decreases the integrity of the lipid bilayer of the fungal membrane (20). Certain amino acid (AA) substitutions in the ligand-binding pocket of SDMs decrease the binding of azole antifungals. For example, two natural AA substitutions, F129 and V291, in Mucormycota SDMs cause intrinsic resistance to short-tail azoles such as fluconazole and voriconazole (21). The T301I substitution in the SDM of A. fumigatus causes intrinsic resistance to fluconazole (22).
Efflux pump activity
Efflux pump activity is a key mechanism of intrinsic resistance. Drugs need to reach a certain level in their cellular targets. Intrinsically resistant fungi exhibit high efflux pump activity, which actively expels antifungal agents from the cell, decreasing their intracellular concentrations and diminishing their efficacy (23). A common efflux system in fungi involves the ATP-binding cassette (ABC) superfamily and major facilitator superfamily (MFS) (23). Fungi express multiple ABCs that are key to their physiology, particularly for the expulsion of toxic metabolites. For example, N. glabratus has 18 ABC-encoding genomes and A. fumigatus has 50 genomes (24,25). In C. auris, four efflux pump encoding genes, Subsequent to Nucleotide Q21 (SNQ21), Subsequent to Nucleotide Q22 (SNQ22), Multidrug Resistance 1 (MDR1) and Candida Drug Resistance 1 (CDR1), contribute to fluconazole resistance (26).
Cell wall and membrane composition changes
Fungal cell wall and membrane composition serve key roles in intrinsic resistance. For example, variations in sterol content affect the binding and efficacy of polyene antifungals such as amphotericin B. Environmental molds such as those from Mucoromycota and Lomentospora spp. have unique cell wall structures that limit the penetration and action of azole antifungals, thereby contributing to their intrinsic resistance (21). Mucoromycota phyla such as Rhizopus and Mucor species have alternative sterol synthesis pathways (21). Duplication of the gene encoding FK506-sensitivity gene (FKS) synthetase, which synthesizes the cell wall component (1,3)-d-glucan, is responsible for the intrinsic resistance of Mucor circinelloides to micafungin (27). Fusarium species also exhibit resistance to several antifungal agents, including azoles, because of their unique cell wall structures, which impede drug penetration, in addition to their robust efflux pump systems (28).
The intrinsic resistance of fungal species highlights the need to develop novel antifungal agents that bypass the resistance mechanisms inherent to these fungi. Additionally, the accurate and timely identification of intrinsically resistant fungal species is key for effective treatment, emphasizing the need for advanced diagnostic tools to identify resistance profiles. Some therapeutic strategies can be employed using alternative drugs or combining them with efflux pump inhibitors or other antifungals that target cell walls.
Antifungal tolerance
Tolerance is a phenomenon distinct from resistance (Table II). Antifungal tolerance refers to the ability of a subpopulation of fungal cells to survive and proliferate in the presence of antifungal agents at concentrations exceeding the established MIC, without exhibiting permanent resistance mechanisms. This tolerance is hypothesized to arise through genetic, physiological or epigenetic adaptations to drugs (29). By contrast, antifungal resistance involves heritable genomic changes that confer a permanent inability to be effectively treated by antifungal agents, typically manifesting as increased MICs (30–32).
However, in practice, tolerance and resistance are not always distinguishable. The MIC within the same fungal species can vary and only strains with considerably different MICs have been recognized as resistant in several studies (33–35). Clinically, patients with infection caused by tolerant strains may initially respond to antifungal therapy but experience a resurgence of symptoms or persistent infection despite appropriate drug levels. This is typically due to the survival of a subpopulation of tolerant cells that can proliferate when drug concentrations fall below the effective levels (12,32,36). By contrast, patients infected with resistant strains typically do not respond to standard antifungal treatments from the outset, as these strains possess inherent mechanisms that allow them to survive at therapeutic concentrations (32,36). Therefore, resistant strains often require higher doses or combination therapy, which can increase the risks of drug toxicity and side effects. Clinicians may need to switch to second-line or less commonly used antifungal agents that may be less effective or have more adverse effects. However, tolerance may require different treatment approaches compared with resistance, such as prolonging the treatment duration to ensure that the surviving fungal cells are eradicated without switching to a second-line treatment.
Genetic analysis and time-kill assays may identify specific mutations associated with resistance, whereas tolerance may not be associated with stable genetic changes but rather transient stress responses (30,37). For example, the presence of mutations in genes associated with drug targets or efflux pumps may indicate resistance, whereas the expression of stress response genes may suggest tolerance (32). In time-kill assays, fungal cells are exposed to antifungal agents over time, and viability is assessed at intervals. A characteristic of tolerance is that cells may initially survive at high drug concentrations but eventually succumb to treatment, whereas resistant strains continue to proliferate despite the presence of the drug (12,38). This method can reveal the dynamics of fungal survival and proliferation in response to antifungal exposure, which may differentiate between transient tolerance and stable resistance.
Acquired resistance
Acquired resistance in fungi refers to the development of resistance mechanisms that enable previously susceptible fungal species to proliferate despite the presence of antifungal drug concentrations that would typically inhibit or kill them. Unlike intrinsic resistance, which is a natural trait of certain species, acquired resistance results from genetic mutations and adaptations that occur in response to antifungal exposure. This resistance poses a considerable challenge in the clinical setting, leading to treatment failure. The development of acquired resistance in fungi involves genetic mechanisms, including point mutations, gene duplication, transposon insertions, aneuploidy and loss of heterozygosity (LOH) (39). In addition to genetic mechanisms, epigenetic mechanisms and phenotypical resistance are also involved. These mechanisms can alter drug targets, enhance drug efflux or modify cellular pathways to decrease the efficacy of antifungal agents (39).
Acquired resistance in fungi has clinical implications, particularly for the treatment of invasive fungal infection. The emergence of drug-resistant strains can lead to treatment failure, prolonged infection and increased healthcare costs (39). The development of acquired resistance can lead to the failure of standard antifungal therapy, necessitating alternative agents that may be less effective or have a greater number of adverse side effects (39). Infection caused by resistant fungal strains is associated with high morbidity and mortality rates because of the limited effectiveness of available antifungal agents (39,40). For example, a national-scale multi-centre cohort study in the Netherlands revealed that infection by azole-resistant A. fumigatus increased mortality by 20–30% compared with azole-susceptible infection (40).
Genetic mechanisms of acquired resistance
Antifungal resistance is a complex and multifaceted phenomenon driven by genetic mechanisms that enable organisms to survive and proliferate despite antifungal treatments. These mechanisms include a range of genetic alterations such as point mutations, gene duplications, transposon insertion, aneuploidy, LOH and hypermutator lineages. Each mechanism contributes to resistance by modifying specific genes or regulatory pathways, enhancing the ability of fungi to withstand antifungal agents and complicating diagnosis and treatment (Table III).
Point mutations
Point mutations are a key genetic mechanism through which fungi acquire resistance to antifungal agents. Point mutations occur at a rate of 1×10−6−1×10−8/cell/generation and involve changes in a single nucleotide base pair within the DNA sequence (41,42). Such alterations lead to modifications in the structure or function of the target protein, thereby diminishing the efficacy of antifungal drugs. Point mutations have clinical implications as they often result in resistance to commonly used antifungal agents, complicating the management of fungal infection.
Point mutations can result in changes in the genetic code, including missense, nonsense and silent mutations. Missense mutations, which result in the substitution of one AA with another in the protein product, alter the structure of the target enzyme or protein, thereby affecting its interaction with antifungal drugs (43,44). Nonsense mutations introduce a premature stop codon into the DNA sequence, producing a truncated and often non-functional protein (43,44). Although silent mutations do not change the AA sequence of a protein, they affect protein folding or stability, potentially influencing drug susceptibility (43,44).
Point mutations confer resistance to antifungal agents in clinically relevant fungal species. One example is the ERG11 gene in C. albicans (Fig. 1A). The ERG11 gene encodes 14α-SDM, an enzyme targeted by azole antifungals such as fluconazole (45). Point mutations in ERG11 lead to AA substitutions that reduce its binding affinity for azole drugs, thereby conferring drug resistance (45). For example, the Y132H mutation in ERG11, which changes a tyrosine to a histidine at position 132, alters the structure of the enzyme and decreases drug binding, resulting in fluconazole resistance (45). Other point mutations in the ERG11 gene of C. albicans (including K143Q, Y205E, A255V, E260V, N435V, G472R and D502E) decrease the ability of fluconazole and voriconazole to inhibit the drug target (45).
Similarly, mutations in the FKS genes, which encode subunits of β-1,3-D-glucan synthase, lead to decreased susceptibility to echinocandin antifungals. In N. glabratus, the S629P mutation in FKS2, in which serine is replaced by proline at position 629, affects the interaction of the enzyme with the drug, thus conferring resistance to echinocandins (46). In A. fumigatus, mutations, such as S678P, in FKS1 confer resistance by altering the drug-binding site of the enzyme (47). In C. albicans, mutations in the gene encoding FKS synthetase, the target of echinocandins, decrease susceptibility to these antifungal agents (48). Another example is the point mutation in the CYP51A gene of A. fumigatus. Point mutations in CYP51A lead to azole resistance, particularly to itraconazole (49). The G54E mutation, which changes glycine to glutamic acid at position 54, markedly decreases the efficacy of itraconazole and other azoles by altering enzyme structure (49).
Among resistance mechanisms, point mutations that target genes, particularly ERG11 and FKS1/FKS2, are the most clinically relevant. These mutations directly impair drug-target binding, resulting in resistance (46,47). Their genomic stability ensures resistance persists in the absence of antifungal pressure. Clinically, such mutations are among the most frequently observed in treatment-refractory infection and are often associated with elevated MICs that render standard antifungal regimens ineffective (45–48,50,51). Consequently, infection caused by fungi harbouring these mutations is harder to treat and requires alternative or combination therapies, highlighting the serious clinical impact.
Gene duplication and transposon insertions
Gene duplication and transposon insertion are key genetic mechanisms that contribute to antifungal resistance. These alterations lead to the increased expression of resistance genes or disruption of regulatory pathways that control drug susceptibility, thereby enhancing the ability of fungi to survive in the presence of antifungal agents. These mechanisms occur at a relatively high frequency of 1×10−3−1×10−4/cell/generation, making them notable drivers of resistance in fungal populations (41,42).
Gene duplication involves the creation of one or more copies of a gene within the genome, leading to the upregulation of genes that confer resistance to antifungal agents. Increased gene expression can enhance the production of proteins that degrade the antifungal drug, modify the drug target or expel the drug from the cell, thereby decreasing its efficacy. A notable example of gene duplication is the MDR1 gene in C. albicans and C. dubliniensis (52,53). The MDR1 gene encodes the MFS transporter involved in drug efflux. Duplication of the MDR1 gene leads to the upregulation of the efflux pump, increasing the expulsion of azole antifungals such as fluconazole from the cell, thereby conferring resistance (52,53). Clinical isolates of C. albicans with duplicated MDR1 genes exhibit high levels of resistance to fluconazole, the most commonly used antifungal (52,53). Similarly, duplication of the ERG11 gene in N. glabratus (Fig. 1B), which encodes 14α-SDM, results in overproduction of the enzyme and is associated with resistance to multiple azole antifungals, making infections difficult to treat (54). An increased amount of the target enzyme can sequester the drug, decreasing its inhibitory effect and leading to drug resistance.
Transposon insertions involve the movement of transposable elements or ‘jumping genes’, within the genome. These elements insert themselves into various genomic locations, disrupting genes or regulatory regions and leading to altered gene expression. Transposon insertions can enhance the expression of resistance genes or disrupt the genes involved in susceptibility to antifungal agents.
The clinical and public health importance of transposon insertions in antifungal resistance has been observed in C. neoformans, which causes 15% of HIV-associated mortalities worldwide (55). The standard treatment for C. neoformans combines rapamycin and FK506 (Fig. 1C) (56). Sequencing of drug target genes revealed C. neoformans LINE-1-like element (CNL1) transposon insertions in an isolate of C. neoformans are resistant to rapamycin and FK506 (56). Cnl1 transposon insertion can cause resistance to 5-flucytosine, which is the standard therapy for cryptococcal meningitis and encephalitis (56).
Aneuploidy
Aneuploidies, defined as the gain or loss of entire chromosomes or large chromosomal segments, are key contributors to antifungal resistance due to their ability to cause widespread changes in gene expression (57,58). Unlike point mutations or gene duplication, these large-scale genomic alterations can upregulate resistance genes or disrupt cell pathways, thereby enhancing fungal survival under drug pressure. Fungi exhibit high genomic plasticity, making aneuploidy a relatively frequent adaptive mechanism. It arises from errors in chromosomal segregation during cell division and typically results in increased expression of drug targets, efflux pumps or other resistance-associated genes (59). This enables rapid and reversible resistance during antifungal treatment.
One of the most studied examples of aneuploidy in antifungal resistance involves C. albicans (60–66). In C. albicans, fluconazole resistance is frequently associated with the formation of an isochromosome consisting of the left arm of chromosome 5, which contains the ERG11 gene (target of fluconazole) and transcriptional activator of efflux pump CDR gene 1 (TAC1) (60–62). This chromosomal alteration increases resistance by enhancing drug target abundance and efflux activity. Similarly, in C. neoformans, duplication of chromosome 1 is associated with fluconazole resistance through upregulation of ERG11 and azole fungal resistance 1 (AFR1), particularly under antifungal stress conditions (Fig. 1D) (67,68).
Aneuploidy in fungal pathogens has notable clinical implications, as it enables rapid gene expression changes that confer drug resistance and complicate treatment. This can lead to treatment failure, prolonged infection and the need for combination or novel antifungal therapy (69). Rapid acquisition of aneuploidy under antifungal pressure allows fungi to adapt quickly, increasing resistance risk during therapy. Therefore, advanced diagnostic techniques, such as karyotyping, comparative genomic hybridization and next-generation sequencing, are key for detecting aneuploidy, guiding treatment and monitoring resistance emergence.
Aneuploidy also helps fungi to mitigate decreased growth and survival due to the acquisition of antifungal resistance. Resistant strains frequently exhibit slower proliferation rates compared with wild-type strains under drug-free conditions. This has been observed, for example, in C. albicans with ERG11 mutations, where the resistant strains demonstrate decreased proliferation rates and virulence in the absence of azole or echinocandin exposure (70). However, in certain cases, compensatory mutations or genomic adaptations through aneuploidy can partially or fully restore fitness, allowing resistant strains to proliferate at rates comparable with wild-type strains even without antifungal agents (63,65). For example, C. albicans strains exhibit distinct transcriptional profiles that lead to altered proliferation and oxidative balance relative to euploid strains (65). Acquisition of aneuploidy in C. albicans can lead to increased fitness during the evolution of antifungal drug resistance. Specifically, the formation of an isochromosome 5L, which amplifies the ERG11 and TAC1 genes, confers fluconazole resistance while also enhancing proliferation rates in the absence of the drug (63).
LOH
LOH is a key genetic mechanism underlying antifungal resistance. LOH refers to mitotic recombination in which one allele at a specific locus is lost, converting heterozygous into homozygous loci (71). Compared with other resistance mechanisms, LOH may occur at higher frequencies under antifungal pressure, particularly in diploid fungi, facilitating rapid adaptation by unmasking recessive resistance-conferring mutations (72). The baseline rates of LOH is 1×10−6−1×10−7/cell division in C. albicans, but these rates can increase ≥285-fold, under antifungal stress such as fluconazole exposure, reaching frequencies of 1×10−3−1×10−4 (72). These elevated rates are markedly higher compared with those of point mutations (~1×10−9) and suggest that LOH is a key driver of rapid adaptation and antifungal resistance in diploid fungal pathogens (72). This genetic alteration can unmask the recessive mutations that confer resistance and enhance the ability of an organism to survive in the presence of antifungal agents. LOH can occur through numerous mechanisms, including mitotic recombination, gene conversion and chromosomal deletion (71). This process is particularly relevant in diploid organisms where heterozygous loci are common.
In diploid fungi, LOH can lead to the unmasking of resistance-conferring mutations that were hidden by a dominant wild-type allele. When a diploid organism loses one copy of a gene, the remaining allele is expressed (73). An organism exhibits a resistant phenotype if the retained allele carries a resistance mutation (73). This allows fungi to adapt quickly to antifungal pressures through advantageous mutations.
An example of the effect of LOH on antifungal resistance is the MDR1 gene in C. albicans (Fig. 1E). The MDR1 gene encodes an MFS transporter involved in drug efflux (74). LOH can lead to the loss of the wild-type allele and retention of a mutant MDR1 allele, resulting in overexpression of the efflux pump and enhanced resistance to azole antifungals (74). Similarly, the LOH in the Δ5,6-desaturase (ERG3P) gene in C. albicans, involved in the ergosterol biosynthesis pathway, causes resistance to amphotericin B (75). Mutations in ERG3 alter the cell membrane composition and LOH can unmask these mutations, leading to decreased susceptibility to amphotericin B and posing treatment challenges (75). Another example is the LOH-mating-type-like (MTL) locus, which causes resistance to azole antifungal agents (76). Single nucleotide polymorphism (SNP) analysis in azole-resistant C. albicans isolates revealed that replacing an MTL locus with a duplicate of the other locus results in two homozygous copies of the MTL locus (76).
LOH of fungal pathogens has clinical implications. LOH can lead to rapid and substantial changes in gene expression and unmasking of resistance mutations that complicate treatment strategies. Advanced diagnostic tools such as comparative genomic hybridization and next-generation sequencing are key for identifying LOH events in clinical isolates, guiding appropriate antifungal therapy and monitoring the emergence of resistance. SNP and Southern blot analyses can also be used to detect common LOH.
Hypermutator lineage
Hypermutator lineages are a notable genetic mechanism contributing to antifungal resistance. Elevated mutation rates characterize these lineages due to defects in DNA repair mechanisms, particularly those involved in mismatch repair (MMR) (77). The increased mutation frequency enhances the genetic diversity within a fungal population, providing a larger pool of mutations from which resistance-conferring changes can arise (77). This mechanism is particularly concerning in clinical settings, as it can lead to the rapid development of resistance to multiple antifungal agents, complicating treatment.
One of the well-documented examples of the hypermutator lineage involves N. glabratus (Fig. 1F) (78–81). Mutations in MutS Homolog 2 (MSH2) gene, a key component of the MMR system, lead to hypermutator phenotypes (78,79). The MSH2 gene is responsible for recognizing and initiating the repair of mismatched nucleotides during DNA replication (78,79). Clinical isolates of N. glabratus with MSH2 mutations exhibit elevated mutation rates that rapidly lead to resistance to multiple antifungal agents, including azoles, echinocandins and polyenes (78,79). These hypermutator strains are challenging to treat because of their ability to quickly adapt to antifungal pressures.
In C. neoformans, hypermutator lineages are associated with mutations in the MSH2 and MutL Homolog 1 genes, which are involved in the MMR pathway. These mutations result in defective repair systems and increased genetic variability (56,82). Hypermutator strains of C. neoformans develop resistance to antifungal agents, such as fluconazole and amphotericin B, more rapidly than non-hypermutator strains (56,82). This rapid adaptation poses challenges to the management of cryptococcal infection, particularly in immunocompromised patients.
Although less commonly reported than C. neoformans and N. glabratus, hypermutator phenotypes have also been observed in A. fumigatus (77,79,83). Mutations in the DNA repair gene MSH lead to increased mutation rates and the rapid emergence of resistance (83). A study of environmental and clinical isolates of A. fumigatus revealed that hypermutator strains quickly develop resistance to azole antifungals, which are commonly used to treat aspergillosis (83). Additionally, genetic instability caused by MSH mutations increases virulence (83). The presence of these strains in clinical settings underscores the need for monitoring and development of alternative therapeutic strategies.
The emergence of hypermutator lineages of fungal pathogens has key clinical implications. These strains rapidly develop resistance to multiple antifungal agents, making infection more difficult to treat and increasing the risk of treatment failure (56,78,79,82,83). The elevated mutation rates in hypermutator lineages lead to a high degree of genetic variability, which can result in the selection of resistance-conferring mutations under antifungal pressure (56,78,79,82,83).
Phenotypical heterogeneity and resistance
Phenotypical heterogeneity refers to the presence of different phenotypes in genetically identical populations of fungal cells. This diversity can affect the susceptibility of fungi to antifungal agents and contribute to the development and persistence of antifungal resistance (10,84–86). Phenotypical heterogeneity allows a subset of the fungal population to survive under adverse conditions such as antifungal treatment, thereby promoting the survival and adaptation of the species (87–90).
Biofilm formation
Phenotypical heterogeneity in fungi arises through numerous mechanisms. A notable example is biofilm formation. Fungal cells in biofilms exist in a sessile, multicellular state and produce an extracellular matrix that protects them from antifungal agents (85,86). This matrix serves as a physical barrier that reduces the effective concentration of the drug reaching fungal cells within the biofilm (85,86). Biofilms are problematic in clinical settings because they are associated with chronic infection and are difficult to eradicate (85,86). Species such as C. albicans can form biofilms on medical devices such as catheters and prosthetic valves, leading to persistent infection (85,86).
Persister fungi
Another mechanism underlying phenotypical heterogeneity involves persister cells. Within a fungal population, a small fraction of cells enter a dormant state, becoming persister cells (10,84). These cells are highly tolerant of antifungal treatment because their reduced metabolic activity makes them less susceptible to drugs targeting active cell processes (10,84). Persister cells do not carry genetic mutations that confer resistance and their tolerance arises from a reversible phenotype (10,84). This makes them challenging as they can survive treatment and later repopulate once the antifungal pressure is removed. Persisters have been extensively studied in Candida spp. and Cryptococcus spp (84,87,88,91). Studies have reported the occurrence of persistent C. neoformans in clinical isolates from patients with cryptococcal meningitis and pneumonia treated with amphotericin B, a standard treatment for cryptococcosis (87,88).
Stress response activation
Fungi activate stress response pathways that alter their phenotype and increase tolerance to antifungal agents. Adaptive responses to stress in human hosts are key for pathogen survival and virulence. For example, the heat shock response lead to the upregulation of chaperone proteins that help the cell cope with the damage caused by antifungal drugs (89). Another example is A. fumigatus, when they are exposed to reactive oxygen species (ROS) produced by immune cells such as macrophages and neutrophils. Analysis of the transcriptomes of A. fumigatus revealed that ROS exposure induces the expression of catalases and superoxide dismutases for survival in an oxidative environment (92). In C. albicans, the activation of the calcineurin pathway in response to azole treatment increases tolerance to the drug, allowing cells to survive otherwise lethal conditions (90).
Epigenetic mechanisms of resistance
Unlike genetic mutations, which alter DNA sequences, epigenetic changes modify gene expression without altering the underlying genetic code. These modifications can be reversible and are influenced by environmental factors, allowing fungi to adapt to antifungal agents rapidly. Understanding these mechanisms is key for the development of novel strategies to combat fungal resistance. Epigenetic changes in fungi primarily involve modifications of histones and DNA methylation, which affect chromatin structure and gene expression. These processes regulate the expression of genes involved in antifungal resistance, including those encoding efflux pumps, drug targets and stress response elements (Table IV).
Histone modification in fungi
Histones are proteins around which DNA is wrapped, forming chromatin. Post-translational histone modification, such as acetylation, methylation, phosphorylation and ubiquitination, influences chromatin structure and gene expression (17,93). Histone acetylation, typically associated with gene activation, is catalysed by histone acetyltransferases and reversed by HDACs (17,93). HDAC inhibitors alter the antifungal resistance by altering the expression of resistance genes.
Histone modifications are among the most studied epigenetic mechanisms in fungi and are linked to antifungal drug resistance, biofilm formation and virulence regulation (94–99). There are seven genes of HDAC in C. neoformans, two of which serve key roles in resistance and virulence. Histone Deacetylase 1 (HDA1) and HDAC genes are required for the survival of C. neoformans in a cultured macrophage model and a mouse model of infection (94,95). Fluconazole works synergistically with HDAC inhibitors in an in vitro infection model of C. neoformans (96). This highlights the fact that HDAC genes modulate the survival and susceptibility of C. neoformans to the antifungal agent.
In C. albicans, three classes of HDAC have been identified: class I, which include proteins similar to yeast Reduced Potassium Dependency 3 (RPD3); class II which consist of proteins related to HDA1; and class III which are NAD+-dependent. SET domain protein 3 (SET3) and Histone Deacetylase of the Rpd3S Complex 2 (HOS2) are HDAC genes that are clinically important in C. albicans, particularly for promoting biofilm formation (97). The deletion of SET3 and HOS2 decreases biofilm formation by C. albicans (97). SET3C complex interacts with master regulator genes of biofilms, such as Negative Regulator Gene 1 (NRG1), Biofilm Regulator Gene 1 (BRG1), TEA/ATTS domain-containing transcription factor 1 (TEC1), Non-Disjunction and Telomere Defect 80 (NDT80) and Regulator of Biofilm 1 (ROB1) (98). HDA1 and RPD3 increase during the early phase of azole resistance in C. albicans and decrease during the later phase (99). HDAC inhibitors increase the sensitivity of C. albicans to azole antifungals by altering the expression of stress response and drug resistance genes (99).
DNA methylation
Although less characterized in fungi compared with higher eukaryotes, DNA methylation serves a key role in transcriptional repression of antifungal target genes and may contribute to stable yet reversible drug resistance. DNA methylation involves the addition of a methyl group to cytosine residues in DNA, typically leading to gene suppression. Although DNA methylation has been well studied in higher eukaryotes, its role in fungi is less clear (100–103). DNA methylation affects the expression of the genes involved in antifungal resistance (93). Changes in methylation patterns can lead to the silencing or activation of genes that help the fungus survive in the presence of antifungal drugs (93).
In C. neoformans, DNA methylation patterns are associated with the resistance to antifungal agents. The DNA methylation in C. neoformans is mediated by the maintenance DNA methyltransferase Dnmt5, which catalyses the deposition of 5-methylcytosine primarily at centromeric and repetitive regions (104). Although this organism lacks a canonical de novo methyltransferase, Dnmt5 maintains heritable methylation patterns across generations (104). Disruption of Dnmt5 causes global hypomethylation, chromosomal instability and altered expression of stress-responsive genes (104). These epigenetic changes may indirectly modulate the expression of genes associated with antifungal resistance, including those encoding efflux pumps and cell wall remodelling proteins. While the spectrum of fungal DNA methyltransferases remains to be characterized, emerging evidence suggests that epigenetic silencing via methylation may contribute to reversible antifungal tolerance (93,104,105).
Chromatin remodelling
Chromatin remodelling allows dynamic control of gene expression in response to antifungal stress, enabling fungi to adjust resistance pathways without permanent genetic mutations. Chromatin remodelling complexes alter the structure of chromatin, making certain regions of the genome more or less accessible to transcription factors and other regulatory proteins. These complexes facilitate the expression of resistance genes by repositioning nucleosomes and exposing regulatory regions of the DNA (17,93). In C. albicans, the chromatin remodelling Switch/sucrose non-fermenting complex 5 (SNF5) subunit is key for tolerance to azoles and amphotericin B under hypoxic stress (106). A study revealed that hypoxia potentiates the antifungal effects of azoles and amphotericin B in C. albicans (106). Remodelling complexes reposition nucleosomes to expose or hide regulatory regions, thereby influencing the expression of resistance genes.
Diagnostic and treatment implications
Diagnostic approaches for resistance detection
Identification of antifungal resistance driven by genetic mechanisms presents diagnostic challenges. The diversity of genetic alterations requires comprehensive and precise diagnostic tools. Traditional methods such as culture-based assays and susceptibility testing may not detect specific genetic changes that confer resistance. Advanced molecular techniques, such as PCR and next-generation and whole-genome sequencing, are essential for detecting specific genetic mutations and alterations (107,108). Techniques such as chromatin immunoprecipitation and bisulfite sequencing are necessary to identify epigenetic changes such as histone modification and DNA methylation patterns that contribute to resistance (109,110). However, these advanced diagnostic tools are expensive and not readily available in all clinical settings, particularly in resource-limited regions. Ensuring accessibility and affordability of these technologies is key for their widespread implementation. Additionally, the time required to perform and analyze advanced diagnostic tests can delay the initiation of appropriate antifungal therapy, potentially worsening patient outcomes. Therefore, rapid diagnostic methods are required to identify resistance mechanisms.
Challenges in antifungal drug development
Development of novel antifungal drugs is challenging due to several interrelated scientific, clinical and economic barriers. Fungi are eukaryotic organisms, sharing key processes with human cells. This limits the availability of pathogen-specific targets and raises the risk of host toxicity (111). Clinically, antifungal drug development must address diverse fungal species, several of which form biofilms, reside in immune-privileged or poorly perfused sites (such as the central nervous system and eyes) and demonstrate intrinsic or acquired resistance to existing therapies (112). Economically, antifungal infections represent a smaller market share than bacterial or viral disease, resulting in lower commercial incentive for pharmaceutical investment (113). In addition, the regulatory pathways for antifungal agents are complex and complicated by the need for robust efficacy data in severely ill and immunocompromised populations, where trial enrolment and design are difficult.
Clinical consequences of resistance mechanisms
Genetic and epigenetic resistance mechanisms complicate treatment of fungal infection. Fungi with hypermutator phenotypes or multiple resistance mechanisms may exhibit resistance to several classes of antifungal drug, thereby limiting the treatment options. This necessitates the use of combination therapy or novel antifungal agents to manage infections effectively. Moreover, epigenetic changes and phenotypical heterogeneity can lead to dynamic and reversible resistance, thereby complicating treatment strategies (17,93). Fungal cells may revert to a resistant state following an initial response to therapy, leading to the recurrence of infection. Higher doses of antifungal agents or prolonged treatment required to overcome resistance can increase the risk of toxicity and adverse side effects (114,115). Balancing efficacy and safety is a key consideration in antifungal therapy.
Strategies to overcome antifungal resistance
Numerous strategies can be used to address the clinical challenges posed by genetic and epigenetic resistance mechanisms. Combinations of antifungal agents with different mechanisms of action can help overcome resistance by simultaneously targeting multiple pathways (116–118). For example, combining azoles with echinocandins or vorinostats may be more effective against biofilm-associated infections than azoles alone (119,120). Continued research and development of antifungal drugs with novel targets are essential to expand the arsenal against resistant fungal strains. Agents that bypass existing resistance mechanisms or target epigenetic modifications hold promise as effective treatments. Developing drugs that modulate epigenetic changes, such as histone deacetylase or DNA methylation inhibitors, can sensitize resistant fungal cells to existing antifungal agents. These approaches offer novel avenues for overcoming resistance.
Numerous strategies are under investigation to address antifungal resistance. Combination therapy (such as azoles with echinocandins or HDAC inhibitors) can enhance efficacy and delay resistance emergence (121). Antifungal classes such as orotomides (for example, olorofim) and glycosylphosphatidylinositol (GPI)-anchor pathway inhibitors (such as fosmanogepix) have demonstrated activity against resistant strains in preclinical and clinical trials (122–124). Adjunctive immunotherapy, including IFN-γ and monoclonal antibodies, offers potential benefit in immunocompromised hosts (125). Antifungal stewardship programs also serve a key role in optimizing antifungal use and minimizing resistance pressure (126).
The identification of novel antifungal drug candidates is achieved through numerous complementary approaches. High-throughput screening of large chemical libraries has led to the discovery of olorofim, a dihydroorotate dehydrogenase inhibitor developed by screening agents targeting fungal pyrimidine biosynthesis, and is in phase III trials against invasive A. fumigatus infections (124,127). Drug repurposing has resulted in identifying sertraline (an antidepressant) with activity against C. neoformans, and tamoxifen, which targets calmodulin in Cryptococcus spp (128,129). In silico modelling and structure-based design have facilitated the development of manogepix (APX001A), which targets GPI-anchored wall transfer protein 1 (GWT1) and has demonstrated broad-spectrum antifungal activity (130). Natural product screening identified ibrexafungerp (SCY-078), a semi-synthetic derivative of enfumafungin, a triterpenoid glycoside (131). Ibrexafungerp inhibits β-1,3-glucan synthase and has been approved for vulvovaginal candidiasis by the U.S. Food and Drug Administration (FDA) in 2021 (131,132). Furthermore, structure-guided drug design based on fungal enzyme crystal structures has supported the optimization of such agents for potency and selectivity (131).
Improving the delivery of antifungal agents to the infection site can increase drug efficacy and decrease resistance. Strategies such as liposomal formulations, nanoparticles and localized delivery systems have been explored to enhance drug delivery (133,134). Developing rapid and accurate diagnostic tools is key for the timely identification of resistance mechanisms and guiding appropriate therapy. Point-of-care diagnostics and streamlined genomic assays help clinicians quickly adapt treatment strategies. By understanding and targeting the underlying mechanisms of resistance, clinicians may improve patient outcomes and effectively manage resistant fungal infection.
To overcome the limitations of conventional antifungal therapy, novel drug delivery systems have been developed to enhance targeted delivery and therapeutic efficacy. Liposomal formulations, such as liposomal amphotericin B, encapsulate antifungal agents within lipid bilayers, enhancing drug solubility, decreasing toxicity and enabling targeted delivery (133,135). A randomized controlled trial in 2022, conducted within resource-limited settings in Africa (Botswana, Malawi, South Africa, Uganda, and Zimbabwe) demonstrated that a single high-dose (10 mg/kg) liposomal amphotericin B regimen, combined with flucytosine and fluconazole, was non-inferior to the conventional 7-day amphotericin B regimen in treating HIV-associated cryptococcal meningitis (136). The simplified regimen markedly decreased adverse events, including nephrotoxicity and anaemia, while maintaining comparable survival outcomes (136). This highlights the clinical efficacy and improved safety profile of liposomal formulations in resource-limited settings, where the use of simplified regimen may decrease the treatment cost.
Nanoparticles can be engineered to respond to stimuli such as pH, temperature or specific enzymes, facilitating drug release at the site of infection, especially where biofilm is involved (134,137). A pre-clinical study demonstrated that ~100 nm diameter liposomal nanoparticles loaded with anidulafungin enhance the solubility and antifungal targeting (138). The formulations were tested in vitro against C. albicans, showing equivalent MICs to free drug for planktonic cells and superior efficacy in disrupting biofilms, decreasing fungal burden by ≥99% (138). In vivo evaluation using a Galleria mellonella infection model demonstrated notably improved survival rates (33–67%) in larvae treated with liposomal formulations compared with 25% in the free drug group, without causing haemolysis, indicating both efficacy and safety (138). A randomized clinical trial evaluated a topical gel containing fluconazole-loaded solid lipid nanoparticles (SLNs) for the treatment of pityriasis versicolor (139). The SLN-based gel demonstrated considerably increased clinical and mycological cure rates compared with conventional fluconazole gel, with enhanced drug retention and patient compliance (139). These findings support the clinical utility of nanoparticle-based topical delivery systems in improving antifungal treatment outcomes.
Localized delivery systems, including hydrogels and coated catheters, enable sustained and concentrated antifungal release at the infection site, particularly for biofilm- or device-related infections (140). In a comparative in vitro biofilm colonization model, central venous catheters (CVCs) and peripherally inserted central catheters impregnated with a novel chlorhexidine-minocycline-rifampin (CHX-M/R) combination were evaluated against resistant bacteria (methicillin-resistant Staphylococcus aureus, vancomycin-resistant enterococci, Pseudomonas aeruginosa) and fungi (C. albicans, N. glabratus) (140). The CHX-M/R-coated catheters completely inhibited biofilm colonization by all tested pathogens at all time points and revealed considerably superior efficacy compared with first-generation M/R catheters, CHX-silver sulfadiazine-treated CVC and uncoated controls (140). The aforementioned studies support the potential of CHX-M/R-coated catheters for sustained, broad-spectrum antimicrobial and antifungal protection, particularly in preventing biofilm-associated device infection.
Overall, the clinical implications of antifungal resistance driven by genetic and epigenetic mechanisms present notable diagnostic and therapeutic challenges. Addressing these challenges requires a multifaceted approach, including the development of advanced diagnostics, novel antifungal agents, combination therapy and strategies targeting epigenetic modifications.
Conclusion
Antifungal resistance is a complex and growing threat that enables fungal pathogens to evade treatment and persist in hostile environments. Antifungal resistance is not a static, mutation-driven process but a dynamic, multilayered phenomenon involving both stable genetic alterations and reversible epigenetic adaptations. The present review highlights key resistance mechanisms, including point mutations, aneuploidy, histone modification and chromatin remodelling and emphasizes their clinical consequences in limiting the efficacy of antifungal therapies. Addressing this challenge requires integrated solutions: rapid molecular diagnostics for early detection, development of novel antifungal agents such as olorofim and fosmanogepix, use of combination and epigenetic-targeting therapies and the implementation of antifungal stewardship programs to preserve drug efficacy. Translating mechanistic insights into diagnostic and therapeutic innovations is key to improve patient outcomes and contain the spread of resistance.
Acknowledgements
The authors would like to thank Ms Rahmati Putri Yaniafari (Nanyang Technological University, Singapore, Singapore) for providing access to several sources of information for this review.
Funding
Funding: No funding was received.
Availability of data and materials
Not applicable.
Authors' contributions
INK, MAE, TIS and YA conceived the study and wrote and revised the manuscript. All authors have read and approved the final manuscript. Data authentication is not applicable.
Ethics approval and consent to participate
Not applicable.
Patient consent for publication
Not applicable.
Competing interests
The authors declare that they have no competing interests.
Use of artificial intelligence tools
During the preparation of this work, artificial intelligence tools were used to improve the readability and language of the manuscript, and subsequently, the authors revised and edited the content produced by the artificial intelligence tools as necessary, taking full responsibility for the ultimate content of the present manuscript.
References
Bongomin F, Gago S, Oladele RO and Denning DW: Global and multi-national prevalence of fungal diseases-estimate precision. J Fungi (Basel). 3:572017. View Article : Google Scholar : PubMed/NCBI | |
Brown GD, Denning DW, Gow NAR, Levitz SM, Netea MG and White TC: Hidden killers: Human fungal infections. Sci Transl Med. 4:165rv13. 2012. View Article : Google Scholar : PubMed/NCBI | |
Denning DW: Global incidence and mortality of severe fungal disease. Lancet Infect Dis. 24:e428–e438. 2024. View Article : Google Scholar : PubMed/NCBI | |
Fisher MC, Hawkins NJ, Sanglard D and Gurr SJ: Worldwide emergence of resistance to antifungal drugs challenges human health and food security. Science. 360:739–742. 2018. View Article : Google Scholar : PubMed/NCBI | |
Enoch DA, Yang H, Aliyu SH and Micallef C: The changing epidemiology of invasive fungal infections. Methods Mol Biol. 1508:17–65. 2017. View Article : Google Scholar | |
Lee Y, Puumala E, Robbins N and Cowen LE: Antifungal drug resistance: Molecular mechanisms in Candida albicans and beyond. Chem Rev. 121:3390–3411. 2020. View Article : Google Scholar : PubMed/NCBI | |
Maligie MA and Selitrennikoff CP: Cryptococcus neoformans resistance to echinocandins:(1, 3) β-glucan synthase activity is sensitive to echinocandins. Antimicrob Agents Chemother. 49:2851–2856. 2005. View Article : Google Scholar : PubMed/NCBI | |
Berkow EL and Lockhart SR: Fluconazole resistance in Candida species: A current perspective. Infect Drug Resist. 10:237–245. 2017. View Article : Google Scholar | |
Pfaller MA, Diekema DJ, Turnidge JD, Castanheira M and Jones RN: Twenty years of the SENTRY antifungal surveillance program: Results for Candida species from 1997–2016. Open Forum Infect Dis. 6 (Suppl 1):S79–S94. 2019. View Article : Google Scholar : PubMed/NCBI | |
Wuyts J, Van Dijck P and Holtappels M: Fungal persister cells: The basis for recalcitrant infections? PLoS Pathog. 14:e10073012018. View Article : Google Scholar : PubMed/NCBI | |
Berman J and Krysan DJ: Drug resistance and tolerance in fungi. Nat Rev Microbiol. 18:319–331. 2020. View Article : Google Scholar | |
Rosenberg AJ, Ene IV, Bibi M, Zakin S, Segal ES, Ziv N, Dahan AM, Colombo AL, Bennett RJ and Berman J: Antifungal tolerance is a subpopulation effect distinct from resistance and is associated with persistent candidemia. Nat Commun. 9:24702018. View Article : Google Scholar : PubMed/NCBI | |
Cowen LE, Sanglard D, Howard SJ, Rogers PD and Perlin DS: Mechanisms of antifungal drug resistance. Cold Spring Harb Perspect Med. 5:a0197522015. View Article : Google Scholar | |
Lee Y, Robbins N and Cowen LE: Molecular mechanisms governing antifungal drug resistance. NPJ Antimicrob Resist. 1:52023. View Article : Google Scholar | |
Revie NM, Iyer KR, Robbins N and Cowen LE: Antifungal drug resistance: Evolution, mechanisms and impact. Curr Opin Microbiol. 45:70–76. 2018. View Article : Google Scholar | |
Shapiro RS, Robbins N and Cowen LE: Regulatory circuitry governing fungal development, drug resistance, and disease. Microbiol Mol Biol Rev. 75:213–267. 2011. View Article : Google Scholar : PubMed/NCBI | |
Patra S, Raney M, Pareek A and Kaur R: Epigenetic regulation of antifungal drug resistance. J Fungi (Basel). 8:8752022. View Article : Google Scholar : PubMed/NCBI | |
Perea S and Patterson TF: Antifungal resistance in pathogenic fungi. Clin Infect Dis. 35:1073–1080. 2002. View Article : Google Scholar : PubMed/NCBI | |
Hosseini P, Keniya MV, Sagatova AA, Toepfer S, Müller C, Tyndall JDA, Klinger A, Fleischer E and Monk BC: The molecular basis of the intrinsic and acquired resistance to azole antifungals in Aspergillus fumigatus. J Fungi (Basel). 10:8202024. View Article : Google Scholar : PubMed/NCBI | |
Rosam K, Monk BC and Lackner M: Sterol 14α-Demethylase ligand-binding pocket-mediated acquired and intrinsic azole resistance in fungal pathogens. J Fungi (Basel). 7:10.3390/jof7010001. 2021. View Article : Google Scholar | |
Caramalho R, Tyndall JDA, Monk BC, Larentis T, Lass-Flörl C and Lackner M: Intrinsic short-tailed azole resistance in mucormycetes is due to an evolutionary conserved aminoacid substitution of the lanosterol 14α-demethylase. Sci Rep. 7:158982017. View Article : Google Scholar : PubMed/NCBI | |
Leonardelli F, Macedo D, Dudiuk C, Cabeza MS, Gamarra S and Garcia-Effron G: Aspergillus fumigatus intrinsic fluconazole resistance is due to the naturally occurring T301I substitution in Cyp51Ap. Antimicrob Agents Chemother. 60:5420–5426. 2016. View Article : Google Scholar : PubMed/NCBI | |
Cannon RD, Lamping E, Holmes AR, Niimi K, Baret PV, Keniya MV, Tanabe K, Niimi M, Goffeau A and Monk BC: Efflux-mediated antifungal drug resistance. Clin Microbiol Rev. 22:291–321. 2009. View Article : Google Scholar : PubMed/NCBI | |
Lamping E, Baret PV, Holmes AR, Monk BC, Goffeau A and Cannon RD: Fungal PDR transporters: Phylogeny, topology, motifs and function. Fungal Genet Biol. 47:127–142. 2010. View Article : Google Scholar | |
Kovalchuk A and Driessen AJM: Phylogenetic analysis of fungal ABC transporters. BMC Genomics. 11:1–21. 2010. View Article : Google Scholar : PubMed/NCBI | |
Chow EWL, Song Y, Chen J, Xu X, Wang J, Chen K, Gao J and Wang Y: The transcription factor Rpn4 activates its own transcription and induces efflux pump expression to confer fluconazole resistance in Candida auris. mBio. 14:e02688–e02623. 2023. View Article : Google Scholar : PubMed/NCBI | |
Garcia A, Huh EY and Lee SC: Serine/Threonine phosphatase calcineurin orchestrates the intrinsic resistance to micafungin in the human-pathogenic fungus mucor circinelloides. Antimicrob Agents Chemother. 67:e00686–e00622. 2023. View Article : Google Scholar : PubMed/NCBI | |
Al-Hatmi AMS, Meis JF and de Hoog GS: Fusarium: Molecular diversity and intrinsic drug resistance. PLoS Pathog. 12:e10054642016. View Article : Google Scholar : PubMed/NCBI | |
Delarze E and Sanglard D: Defining the frontiers between antifungal resistance, tolerance and the concept of persistence. Drug Resist Updat. 23:12–19. 2015. View Article : Google Scholar | |
Arastehfar A, Lass-Flörl C, Garcia-Rubio R, Daneshnia F, İlkit M, Boekhout T, Gabaldon T and Perlin DS: The quiet and underappreciated rise of drug-resistant invasive fungal pathogens. J Fungi (Basel). 6:1382020. View Article : Google Scholar : PubMed/NCBI | |
Delarze E, Brandt L, Trachsel E, Patxot M, Pralong C, Maranzano F, Chauvel M, Legrand M, Znaidi S, Bougnoux ME, et al: Identification and characterization of mediators of fluconazole tolerance in Candida albicans. Front Microbiol. 11:5911402020. View Article : Google Scholar | |
Koohi SR, Shankarnarayan SA, Galon CM and Charlebois DA: Identification and elimination of antifungal tolerance in Candida auris. Biomedicines. 11:8982023. View Article : Google Scholar : PubMed/NCBI | |
Delma FZ, Melchers WJG, Verweij PE and Buil JB: Wild-type MIC distributions and epidemiological cutoff values for 5-flucytosine and Candida species as determined by EUCAST broth microdilution. JAC Antimicrob Resist. 6:dlae1532024. View Article : Google Scholar | |
Pfaller MA, Boyken L, Hollis RJ, Kroeger J, Messer SA, Tendolkar S, Jones RN, Turnidge J and Diekema DJ: Wild-type MIC distributions and epidemiological cutoff values for the echinocandins and Candida spp. J Clin Microbiol. 48:52–56. 2010. View Article : Google Scholar | |
Espinel-Ingroff A, Colombo AL, Cordoba S, Dufresne PJ, Fuller J, Ghannoum M, Gonzalez GM, Guarro J, Kidd SE, Meis JF, et al: International evaluation of MIC distributions and epidemiological cutoff value (ECV) definitions for fusarium species identified by molecular methods for the CLSI broth microdilution method. Antimicrob Agents Chemother. 60:1079–1084. 2016. View Article : Google Scholar : PubMed/NCBI | |
Levinson T, Dahan A, Novikov A, Paran Y, Berman J and Ben-Ami R: Impact of tolerance to fluconazole on treatment response in Candida albicans bloodstream infection. Mycoses. 64:78–85. 2021. View Article : Google Scholar : PubMed/NCBI | |
Yu S, Paderu P, Lee A, Eirekat S, Healey KR, Chen L, Perlin DS and Zhao Y: Histone acetylation regulator Gcn5 mediates drug resistance and virulence of Candida Glabrata. Microbiol Spectr. 10:e00963222022. View Article : Google Scholar : PubMed/NCBI | |
Garnaud C, García-Oliver E, Wang Y, Maubon D, Bailly S, Despinasse Q, Champleboux M, Govin J and Cornet M: The rim pathway mediates antifungal tolerance in Candida albicans through newly identified Rim101 transcriptional targets, including Hsp90 and Ipt1. Antimicrob Agents Chemother. 62:e01785–e01717. 2018. View Article : Google Scholar : PubMed/NCBI | |
Wiederhold NP: Antifungal resistance: Current trends and future strategies to combat. Infect Drug Resist. 10:249–259. 2017. View Article : Google Scholar | |
Lestrade PPA, Buil JB, van Der Beek MT, Kuijper EJ, van Dijk K, Kampinga GA, Rijnders BJA, Vonk AG, de Greeff SC, Schoffelen AF, et al: Paradoxal trends in azole-resistant Aspergillus fumigatus in a national multicenter surveillance program, the Netherlands, 2013–2018. Emerg Infect Dis. 26:1447–1455. 2020. View Article : Google Scholar : PubMed/NCBI | |
Edwards HM and Rhodes J: Accounting for the biological complexity of pathogenic fungi in phylogenetic dating. J Fungi (Basel). 7:6612021. View Article : Google Scholar : PubMed/NCBI | |
White TC, Marr KA and Bowden RA: Clinical, cellular, and molecular factors that contribute to antifungal drug resistance. Clin Microbiol Re. 11:382–402. 1998. View Article : Google Scholar | |
Gladyshev E: Repeat-Induced Point Mutation and Other Genome Defense Mechanisms in Fungi. Microbiol Spectr. 5:102017. View Article : Google Scholar | |
Hane JK, Williams AH, Taranto AP, Solomon PS and Oliver RP: Repeat-induced point mutation: A fungal-specific, endogenous mutagenesis process. Genet Transform Syst Fungi. 2:55–68. 2015. View Article : Google Scholar | |
Xiang MJ, Liu JY, Ni PH, Wang S, Shi C, Wei B, Ni YX and Ge HL: Erg11 mutations associated with azole resistance in clinical isolates of Candida albicans. FEMS Yeast Res. 13:386–393. 2013. View Article : Google Scholar : PubMed/NCBI | |
Biswas C, Marcelino VR, Van Hal S, Halliday C, Martinez E, Wang Q, Kidd S, Kennedy K, Marriott D, Morrissey CO, et al: Whole genome sequencing of Australian Candida glabrata isolates reveals genetic diversity and novel sequence types. Front Microbiol. 9:29462018. View Article : Google Scholar | |
Rocha EMF, Garcia-Effron G, Park S and Perlin DS: A Ser678Pro substitution in Fks1p confers resistance to echinocandin drugs in Aspergillus fumigatus. Antimicrob Agents Chemother. 51:4174–4176. 2007. View Article : Google Scholar : PubMed/NCBI | |
Lackner M, Tscherner M, Schaller M, Kuchler K, Mair C, Sartori B, Istel F, Arendrup MC and Lass-Flörl C: Positions and numbers of FKS mutations in Candida albicans selectively influence in vitro and in vivo susceptibilities to echinocandin treatment. Antimicrob Agents Chemother. 58:3626–3635. 2014. View Article : Google Scholar : PubMed/NCBI | |
Diaz-Guerra TM, Mellado E, Cuenca-Estrella M and Rodriguez-Tudela JL: A point mutation in the 14α-Sterol demethylase gene cyp51A contributes to itraconazole resistance in Aspergillus fumigatus. Antimicrob Agents Chemother. 47:1120–1124. 2003. View Article : Google Scholar : PubMed/NCBI | |
Katiyar SK, Alastruey-Izquierdo A, Healey KR, Johnson ME, Perlin DS and Edlind TD: Fks1 and Fks2 are functionally redundant but differentially regulated in Candida glabrata: Implications for echinocandin resistance. Antimicrob Agents Chemother. 56:6304–6309. 2012. View Article : Google Scholar : PubMed/NCBI | |
Katiyar SK, Alastruey-Izquierdo A, Healey KR, Johnson ME, Perlin DS and Edlind TD: Contribution of clinically derived mutations in ERG11 to azole resistance in Candida albicans. Antimicrob Agents Chemother. 59:450–460. 2014. View Article : Google Scholar : PubMed/NCBI | |
Wirsching S, Moran GP, Sullivan DJ, Coleman DC and Morschhäuser J: MDR1-mediated drug resistance in Candida dubliniensis. Antimicrob Agents Chemother. 45:3416–3421. 2001. View Article : Google Scholar : PubMed/NCBI | |
Morschhäuser J, Barker KS, Liu TT, Blaß-Warmuth J, Homayouni R and Rogers PD: The transcription factor Mrr1p controls expression of the MDR1 efflux pump and mediates multidrug resistance in Candida albicans. PLoS Pathog. 3:e1642007. View Article : Google Scholar | |
Abbes S, Mary C, Sellami H, Michel-Nguyen A, Ayadi A and Ranque S: Interactions between copy number and expression level of genes involved in fluconazole resistance in Candida glabrata. Front Cell Infect Microbiol. 3:742013. View Article : Google Scholar | |
Rajasingham R, Smith RM, Park BJ, Jarvis JN, Govender NP, Chiller TM, Denning DW, Loyse A and Boulware DR: Global burden of disease of HIV-associated cryptococcal meningitis: An updated analysis. Lancet Infect Dis. 17:873–881. 2017. View Article : Google Scholar : PubMed/NCBI | |
Priest SJ, Yadav V, Roth C, Dahlmann TA, Kück U, Magwene PM and Heitman J: Uncontrolled transposition following RNAi loss causes hypermutation and antifungal drug resistance in clinical isolates of Cryptococcus neoformans. Nat Microbiol. 7:1239–1251. 2022. View Article : Google Scholar | |
Torres EM, Sokolsky T, Tucker CM, Chan LY, Boselli M, Dunham MJ and Amon A: Effects of aneuploidy on cellular physiology and cell division in haploid yeast. Science. 317:916–924. 2007. View Article : Google Scholar : PubMed/NCBI | |
Williams BR, Prabhu VR, Hunter KE, Glazier CM, Whittaker CA, Housman DE and Amon A: Aneuploidy affects proliferation and spontaneous immortalization in mammalian cells. Science. 322:703–709. 2008. View Article : Google Scholar : PubMed/NCBI | |
Tsai HJ and Nelliat A: A double-edged sword: Aneuploidy is a prevalent strategy in fungal adaptation. Genes (Basel). 10:7872019. View Article : Google Scholar : PubMed/NCBI | |
Selmecki A, Forche A and Berman J: Aneuploidy and isochromosome formation in drug-resistant Candida albicans. Science. 313:367–370. 2006. View Article : Google Scholar : PubMed/NCBI | |
Ji H, Zhang W, Zhou Y, Zhang M, Zhu J, Song Y and Lü J: A three-dimensional model of lanosterol 14α-demethylase of Candida albicans and its interaction with azole antifungals. J Med Chem. 43:2493–2505. 2000. View Article : Google Scholar : PubMed/NCBI | |
Coste AT, Karababa M, Ischer F, Bille J and Sanglard D: TAC1, transcriptional activator of CDR genes, is a new transcription factor involved in the regulation of Candida albicans ABC transporters CDR1 and CDR2. Eukaryot Cell. 3:1639–1652. 2004. View Article : Google Scholar | |
Selmecki AM, Dulmage K, Cowen LE, Anderson JB and Berman J: Acquisition of aneuploidy provides increased fitness during the evolution of antifungal drug resistance. PLOS Genet. 5:e10007052009. View Article : Google Scholar | |
Ford CB, Funt JM, Abbey D, Issi L, Guiducci C, Martinez DA, Delorey T, Li BY, White TC, Cuomo C, et al: The evolution of drug resistance in clinical isolates of Candida albicans. Elife. 4:e006622015. View Article : Google Scholar : PubMed/NCBI | |
Mackey AI, Fillinger RJ, Hendricks PS, Thomson GJ, Cuomo CA, Bennett RJ and Anderson MZ: Aneuploidy confers a unique transcriptional and phenotypic profile to Candida albicans. Nat Commun. 16:32872025. View Article : Google Scholar : PubMed/NCBI | |
Sun LL, Li H, Yan TH, Fang T, Wu H, Cao YB, Lu H, Jiang YY and Yang F: Aneuploidy mediates rapid adaptation to a subinhibitory amount of fluconazole in Candida albicans. Microbiol Spectr. 11:e03016–e03022. 2023. View Article : Google Scholar : PubMed/NCBI | |
Sionov E, Lee H, Chang YC and Kwon-Chung KJ: Cryptococcus neoformans overcomes stress of azole drugs by formation of disomy in specific multiple chromosomes. PLoS Pathog. 6:e10008482010. View Article : Google Scholar : PubMed/NCBI | |
Zhang Z, Sun L, Fu B, Deng J, Jia C, Miao M, Yang F, Cao YB and Yan TH: Aneuploidy underlies brefeldin A-induced antifungal drug resistance in Cryptococcus neoformans. Front Cell Infect Microbiol. 14:13977242024. View Article : Google Scholar | |
Hu G, Wang J, Choi J, Jung WH, Liu I, Litvintseva AP, Bicanic T, Aurora R, Mitchell TG, Perfect JR and Kronstad JW: Variation in chromosome copy number influences the virulence of Cryptococcus neoformans and occurs in isolates from AIDS patients. BMC Genomics. 12:1–19. 2011. View Article : Google Scholar | |
Sasse C, Dunkel N, Schäfer T, Schneider S, Dierolf F, Ohlsen K and Morschhäuser J: The stepwise acquisition of fluconazole resistance mutations causes a gradual loss of fitness in andida albicans. Mol Microbiol. 86:539–556. 2012. View Article : Google Scholar | |
Heil CS: Loss of heterozygosity and its importance in evolution. J Mol Evol. 91:369–377. 2023. View Article : Google Scholar | |
Forche A, Abbey D, Pisithkul T, Weinzierl MA, Ringstrom T, Bruck D, Petersen K and Berman J: Stress alters rates and types of loss of heterozygosity in Candida albicans. mBio. 2:e00129–e00111. 2011. View Article : Google Scholar : PubMed/NCBI | |
Bennett RJ, Forche A and Berman J: Rapid mechanisms for generating genome diversity: Whole ploidy shifts, aneuploidy, and loss of heterozygosity. Cold Spring Harb Perspect Med. 4:a0196042014. View Article : Google Scholar : PubMed/NCBI | |
White TC: Increased mRNA levels of ERG16, CDR, and MDR1 correlate with increases in azole resistance in Candida albicans isolates from a patient infected with human immunodeficiency virus. Antimicrob Agents Chemother. 41:1482–1487. 1997. View Article : Google Scholar : PubMed/NCBI | |
Luna-Tapia A, Willems HME, Parker JE, Tournu H, Barker KS, Nishimoto AT, Rogers PD, Kelly SL, Peters BM and Palmer GE: Loss of Upc2p-inducible ERG3 transcription is sufficient to confer niche-specific azole resistance without compromising Candida albicans pathogenicity. mBio. 9:e00225–e00218. 2018. View Article : Google Scholar : PubMed/NCBI | |
Rustad TR, Stevens DA, Pfaller MA and White TC: Homozygosity at the Candida albicans MTL locus associated with azole resistance. Microbiology. 148:1061–1072. 2002. View Article : Google Scholar : PubMed/NCBI | |
Gambhir N, Harris SD and Everhart SE: Evolutionary significance of fungal hypermutators: Lessons learned from clinical strains and implications for fungal plant pathogens. mSphere. 7:e00087–e00022. 2022. View Article : Google Scholar : PubMed/NCBI | |
Healey KR, Ortigosa CJ, Shor E and Perlin DS: Genetic drivers of multidrug resistance in Candida glabrata. Front Microbiol. 7:19952016. View Article : Google Scholar | |
Healey KR, Zhao Y, Perez WB, Lockhart SR, Sobel JD, Farmakiotis D, Kontoyiannis DP, Sanglard D, Taj-Aldeen SJ, Alexander BD, et al: Prevalent mutator genotype identified in fungal pathogen Candida glabrata promotes multi-drug resistance. Nat Commun. 7:111282016. View Article : Google Scholar : PubMed/NCBI | |
Helmstetter N, Chybowska AD, Delaney C, Da Silva Dantas A, Gifford H, Wacker T, Munro C, Warris A, Jones B, Cuomo CA, et al: Population genetics and microevolution of clinical Candida glabrata reveals recombinant sequence types and hyper-variation within mitochondrial genomes, virulence genes, and drug targets. Genetics. 221:iyac0312022. View Article : Google Scholar : PubMed/NCBI | |
Vale-Silva L, Beaudoing E, Tran VDT and Sanglard D: Comparative genomics of two sequential Candida glabrata clinical isolates. G3 (Bethesda). 7:2413–2426. 2017. View Article : Google Scholar : PubMed/NCBI | |
Boyce KJ, Wang Y, Verma S, Shakya VPS, Xue C and Idnurm A: Mismatch repair of DNA replication errors contributes to microevolution in the pathogenic fungus Cryptococcus neoformans. mBio. 8:e00595–e00517. 2017. View Article : Google Scholar : PubMed/NCBI | |
Dos Reis TF, Silva LP, de Castro PA, do Carmo RA, Marini MM, da Silveira JF, Ferreira BH, Rodrigues F, Lind AL, Rokas A and Goldman GH: The Aspergillus fumigatus mismatch repair MSH2 homolog is important for virulence and azole resistance. mSphere. 4:e00416–e00419. 2019. View Article : Google Scholar : PubMed/NCBI | |
Ke W, Xie Y, Chen Y, Ding H, Ye L, Qiu H, Li H, Zhang L, Chen L, Tian X, et al: Fungicide-tolerant persister formation during cryptococcal pulmonary infection. Cell Host Microbe. 32:276–289. 2024. View Article : Google Scholar : PubMed/NCBI | |
Silva S, Rodrigues CF, Araújo D, Rodrigues ME and Henriques M: Candida Species Biofilms' antifungal resistance. J Fungi (Basel). 3:82017. View Article : Google Scholar : PubMed/NCBI | |
Massey J, Zarnowski R and Andes D: Role of the extracellular matrix in Candida biofilm antifungal resistance. FEMS Microbiol Rev. 47:fuad0592023. View Article : Google Scholar : PubMed/NCBI | |
Hommel B, Sturny-Leclère A, Volant S, Veluppillai N, Duchateau M, Yu CH, Hourdel V, Varet H, Matondo M, Perfect JR, et al: Cryptococcus neoformans resists to drastic conditions by switching to viable but non-culturable cell phenotype. PLoS Pathog. 15:e10079452019. View Article : Google Scholar : PubMed/NCBI | |
Alanio A, Vernel-Pauillac F, Sturny-Leclère A and Dromer F, Alanio A, Vernel-Pauillac F and Dromer F: Cryptococcus neoformans host adaptation: Toward biological evidence of dormancy. mBio. 6:e02580–e02514. 2015. View Article : Google Scholar : PubMed/NCBI | |
Hayes BME, Anderson MA, Traven A, van der Weerden NL and Bleackley MR: Activation of stress signalling pathways enhances tolerance of fungi to chemical fungicides and antifungal proteins. Cell Mol life Sci. 71:2651–2666. 2014. View Article : Google Scholar | |
Cruz MC, Goldstein AL, Blankenship JR, Del Poeta M, Davis D, Cardenas ME, Perfect JR, McCusker JH and Heitman J: Calcineurin is essential for survival during membrane stress in Candida albicans. EMBO J. 21:546–559. 2002. View Article : Google Scholar : PubMed/NCBI | |
LaFleur MD, Kumamoto CA and Lewis K: Candida albicans biofilms produce antifungal-tolerant persister cells. Antimicrob Agents Chemother. 50:3839–3846. 2006. View Article : Google Scholar : PubMed/NCBI | |
Fréalle E, Aliouat-Denis CM, Delhaes L, Hot D and Dei-Cas E: Transcriptomic insights into the oxidative response of stress-exposed Aspergillus fumigatus. Curr Pharm Des. 19:3713–3737. 2013. View Article : Google Scholar | |
Chang Z, Yadav V, Lee SC and Heitman J: Epigenetic mechanisms of drug resistance in fungi. Fungal Genet Biol. 132:1032532019. View Article : Google Scholar | |
Brandao FAS, Derengowski LS, Albuquerque P, Nicola AM, Silva-Pereira I and Poças-Fonseca MJ: Histone deacetylases inhibitors effects on Cryptococcus neoformans major virulence phenotypes. Virulence. 6:618–630. 2015. View Article : Google Scholar : PubMed/NCBI | |
Brandão F, Esher SK, Ost KS, Pianalto K, Nichols CB, Fernandes L, Bocca AL, Poças-Fonseca MJ and Alspaugh JA: HDAC genes play distinct and redundant roles in Cryptococcus neoformans virulence. Sci Rep. 8:52092018. View Article : Google Scholar | |
Ranjan K, Brandão F, Morais JAV, Muehlmann LA, Silva-Pereira I, Bocca AL, Matos LF and Poças-Fonseca MJ: The role of Cryptococcus neoformans histone deacetylase genes in the response to antifungal drugs, epigenetic modulators and to photodynamic therapy mediated by an aluminium phthalocyanine chloride nanoemulsion in vitro. J Photochem Photobiol B. 216:1121312021. View Article : Google Scholar : PubMed/NCBI | |
Nobile CJ, Fox EP, Nett JE, Sorrells TR, Mitrovich QM, Hernday AD, Tuch BB, Andes DR and Johnson AD: A recently evolved transcriptional network controls biofilm development in Candida albicans. Cell. 148:126–138. 2012. View Article : Google Scholar | |
Uppuluri P, Pierce CG, Thomas DP, Bubeck SS, Saville SP and Lopez-Ribot JL: The transcriptional regulator Nrg1p controls Candida albicans biofilm formation and dispersion. Eukaryot Cell. 9:1531–1537. 2010. View Article : Google Scholar | |
Li X, Cai Q, Mei H, Zhou X, Shen Y, Li D and Liu W: The Rpd3/Hda1 family of histone deacetylases regulates azole resistance in Candida albicans. J Antimicrob Chemother. 70:1993–2003. 2015. View Article : Google Scholar : PubMed/NCBI | |
Freitag M: Histone methylation by SET domain proteins in fungi. Annu Rev Microbiol. 71:413–439. 2017. View Article : Google Scholar | |
Honda S, Bicocca VT, Gessaman JD, Rountree MR, Yokoyama A, Yu EY, Selker JM and Selker EU: Dual chromatin recognition by the histone deacetylase complex HCHC is required for proper DNA methylation in Neurospora crassa. Proc Natl Acad Sci USA. 113:E6135–E6144. 2016. View Article : Google Scholar : PubMed/NCBI | |
Nai YS, Huang YC, Yen MR and Chen PY: Diversity of fungal DNA methyltransferases and their association with DNA methylation patterns. Front Microbiol. 11:6169222021. View Article : Google Scholar | |
Jeon J, Choi J, Lee GW, Park SY, Huh A, Dean RA and Lee YH: Genome-wide profiling of DNA methylation provides insights into epigenetic regulation of fungal development in a plant pathogenic fungus, Magnaporthe oryzae. Sci Rep. 5:85672015. View Article : Google Scholar : PubMed/NCBI | |
Catania S, Dumesic PA, Pimentel H, Nasif A, Stoddard CI, Burke JE, Diedrich JK, Cook S, Shea T, Geinger E, et al: Evolutionary persistence of DNA methylation for millions of years after ancient loss of a de novo methyltransferase. Cell. 180:263–277. 2020. View Article : Google Scholar | |
Baker KM, Hoda S, Saha D, Gregor JB, Georgescu L, Serratore ND, Zhang Y, Cheng L, Lanman NA, Briggs SD, et al: The Set1 histone H3K4 methyltransferase contributes to azole susceptibility in a species-specific manner by differentially altering the expression of drug efflux pumps and the ergosterol gene pathway. Antimicrob Agents Chemother. 66:e02250–e02221. 2022. View Article : Google Scholar : PubMed/NCBI | |
Khemiri I, Tebbji F, Burgain A and Sellam A: Regulation of copper uptake by the SWI/SNF chromatin remodeling complex in Candida albicans affects susceptibility to antifungal and oxidative stresses under hypoxia. FEMS Yeast Res. 24:foae0182024. View Article : Google Scholar : PubMed/NCBI | |
Salem-Bango Z, Price TK, Chan JL, Chandrasekaran S, Garner OB and Yang S: Fungal whole-genome sequencing for species identification: From test development to clinical utilization. J Fungi (Basel). 9:1832023. View Article : Google Scholar : PubMed/NCBI | |
Jiang S, Chen Y, Han S, Lv L and Li L: Next-generation sequencing applications for the study of fungal pathogens. Microorganisms. 10:18822022. View Article : Google Scholar : PubMed/NCBI | |
Liu SY, Lin JQ, Wu HL, Wang CC, Huang SJ, Luo YF, Sun JH, Zhou JX, Yan SJ, He JG, et al: Bisulfite sequencing reveals that Aspergillus flavus holds a hollow in DNA methylation. PLoS One. 7:e303492012. View Article : Google Scholar : PubMed/NCBI | |
Tan K and Wong KH: RNA polymerase II ChIP-seq-a powerful and highly affordable method for studying fungal genomics and physiology. Biophys Rev. 11:79–82. 2019. View Article : Google Scholar : PubMed/NCBI | |
Roemer T and Krysan DJ: Antifungal drug development: Challenges, unmet clinical needs, and new approaches. Cold Spring Harb Perspect Med. 4:a0197032014. View Article : Google Scholar : PubMed/NCBI | |
Fisher MC, Alastruey-Izquierdo A, Berman J, Bicanic T, Bignell E, Bowyer P, Bromley M, Brüggemann R, Garber G, Cornely OA, et al: Tackling the emerging threat of antifungal resistance to human health. Nat Rev Microbiol. 20:557–571. 2022. View Article : Google Scholar | |
Grand View Research, . Antifungal drugs market size, share & growth report 2030 [Internet]. 2025.Available from:. https://www.grandviewresearch.com/industry-analysis/antifungal-drugs-market | |
Pyrpasopoulou A, Iosifidis E, Antachopoulos C and Roilides E: Antifungal drug dosing adjustment in critical patients with invasive fungal infections. J Emerg Crit Care Med. 3:10.21037/jeccm.2019.08.01. 2019. View Article : Google Scholar | |
Baracaldo-Santamaría D, Cala-Garcia JD, Medina-Rincón GJ, Rojas-Rodriguez LC and Calderon-Ospina CA: Therapeutic drug monitoring of antifungal agents in critically ill patients: Is there a need for dose optimisation? Antibiotics (Basel). 11:6452022. View Article : Google Scholar | |
Glampedakis E, Coste AT, Aruanno M, Bachmann D, Delarze E, Erard V and Lamoth F: Efficacy of antifungal monotherapies and combinations against Aspergillus calidoustus. Antimicrob Agents Chemother. 62:e01137–e01118. 2018. View Article : Google Scholar : PubMed/NCBI | |
Rieger CT, Ostermann H, Kolb HJ, Fiegl M, Huppmann S, Morgenstern N and Tischer J: A clinical cohort trial of antifungal combination therapy: Efficacy and toxicity in haematological cancer patients. Ann Hematol. 87:915–922. 2008. View Article : Google Scholar | |
Candoni A, Caira M, Cesaro S, Busca A, Giacchino M, Fanci R, Delia M, Nosari A, Bonini A, Cattaneo C, et al: Multicentre surveillance study on feasibility, safety and efficacy of antifungal combination therapy for proven or probable invasive fungal diseases in haematological patients: the SEIFEM real-life combo study. Mycoses. 57:342–350. 2014. View Article : Google Scholar : PubMed/NCBI | |
Tu B, Yin G and Li H: Synergistic effects of vorinostat (SAHA) and azoles against Aspergillus species and their biofilms. BMC Microbiol. 20:1–7. 2020. View Article : Google Scholar | |
Rodrigues CF, Alves DF and Henriques M: Combination of Posaconazole and Amphotericin B in the treatment of Candida glabrata biofilms. Microorganisms. 6:1232018. View Article : Google Scholar : PubMed/NCBI | |
Vitale RG: Role of antifungal combinations in difficult to treat Candida infections. J Fungi (Basel). 7:7312021. View Article : Google Scholar : PubMed/NCBI | |
Fernandes CM, Dasilva D, Haranahalli K, McCarthy JB, Mallamo J, Ojima I and Del Poeta M: The future of antifungal drug therapy: Novel compounds and targets. Antimicrob Agents Chemother. 65:e01719–e01720. 2021. View Article : Google Scholar : PubMed/NCBI | |
Hodges MR, Tawadrous M, Cornely OA, Thompson GR III, Slavin MA, Maertens JA, Dadwal SS, Rahav G, Hazel S, Almas M, et al: Fosmanogepix for the treatment of invasive mold diseases caused by Aspergillus species and rare molds: A phase 2, open-label study (AEGIS). Clin Infect Dis. 9:ciaf1852025. View Article : Google Scholar | |
Oliver JD, Sibley GEM, Beckmann N, Dobb KS, Slater MJ, McEntee L, du Pré S, Livermore J, Bromley MJ, Wiederhold NP, et al: F901318 represents a novel class of antifungal drug that inhibits dihydroorotate dehydrogenase. Proc Natl Acad Sci USA. 113:12809–12814. 2016. View Article : Google Scholar : PubMed/NCBI | |
Tawfik DM, Dereux C, Tremblay JA, Boibieux A, Braye F, Cazauran JB, Rabodonirina M, Cerrato E, Guichard A, Venet F, et al: Interferon gamma as an immune modulating adjunct therapy for invasive mucormycosis after severe burn-A case report. Front Immunol. 13:8836382022. View Article : Google Scholar | |
Albahar F, Alhamad H, Assab MA, Abu-Farha R, Alawi L and Khaleel S: The impact of antifungal stewardship on clinical and performance measures: A global systematic review. Trop Med Infect Dis. 9:82023. View Article : Google Scholar : PubMed/NCBI | |
Kühbacher A, Birch M, Oliver JD and Gsaller F: Anti-Aspergillus activities of olorofim at sub-MIC levels during early-stage growth. Microbiol Spectr. 12:e03304–e03323. 2024. View Article : Google Scholar | |
Rhein J, Hullsiek KH, Tugume L, Nuwagira E, Mpoza E, Evans EE, Kiggundu R, Pastick KA, Ssebambulidde K, Akampurira A, et al: Adjunctive sertraline for HIV-associated cryptococcal meningitis: A randomised, placebo-controlled, double-blind phase 3 trial. Lancet Infect Dis. 19:843–851. 2019. View Article : Google Scholar : PubMed/NCBI | |
Liu Q, Guo X, Jiang G, Wu G, Miao H, Liu K, Chen S, Sakamoto N, Kuno T, Yao F and Fang Y: NADPH-cytochrome P450 reductase Ccr1 is a target of tamoxifen and participates in its antifungal activity via regulating cell wall integrity in fission yeast. Antimicrob Agents Chemother. 64:e00079–e00072. 2020. View Article : Google Scholar : PubMed/NCBI | |
Dai X, Liu X, Li J, Chen H, Yan C, Li Y, Liu H, Deng D and Wang X: Structural insights into the inhibition mechanism of fungal GWT1 by manogepix. Nat Commun. 15:91942024. View Article : Google Scholar : PubMed/NCBI | |
Schwebke JR, Sobel R, Gersten JK, Sussman SA, Lederman SN, Jacobs MA, Chappell BT, Weinstein DL, Moffett AH, Azie NE, et al: Ibrexafungerp versus placebo for vulvovaginal candidiasis treatment: A phase 3, randomized, controlled superiority trial (VANISH 303). Clin Infect Dis. 74:1979–1985. 2022. View Article : Google Scholar : PubMed/NCBI | |
(FDA) USF and DA, . Drug trials snapshots: brexafemme [Internet]. FDA; 2023, Available from:. https://www.fda.gov/drugs/drug-approvals-and-databases/drug-trials-snapshots-brexafemme | |
Firooz A, Nafisi S and Maibach HI: Novel drug delivery strategies for improving econazole antifungal action. Int J Pharm. 495:599–607. 2015. View Article : Google Scholar : PubMed/NCBI | |
Nami S, Aghebati-Maleki A and Aghebati-Maleki L: Current applications and prospects of nanoparticles for antifungal drug delivery. EXCLI J. 20:562–584. 2021.PubMed/NCBI | |
Stone NRH, Bicanic T, Salim R and Hope W: Liposomal amphotericin B (AmBisome®): A review of the pharmacokinetics, pharmacodynamics, clinical experience and future directions. Drugs. 76:485–500. 2016. View Article : Google Scholar : PubMed/NCBI | |
Jarvis JN, Lawrence DS, Meya DB, Kagimu E, Kasibante J, Mpoza E, Rutakingirwa MK, Ssebambulidde K, Tugume L, Rhein J, et al: Single-dose liposomal amphotericin B treatment for cryptococcal meningitis. N Engl J Med. 386:1109–1120. 2022. View Article : Google Scholar : PubMed/NCBI | |
Empitu MA, Kadariswantiningsih IN and Shakri NM: Pharmacological strategies for targeting biofilms in otorhinolaryngologic infections and overcoming antimicrobial resistance. Biomed Rep. 22:952025. View Article : Google Scholar : PubMed/NCBI | |
Vera-González N, Bailey-Hytholt CM, Langlois L, de Camargo Ribeiro F, de Souza Santos EL, Junqueira JC and Shukla A: Anidulafungin liposome nanoparticles exhibit antifungal activity against planktonic and biofilm Candida albicans. J Biomed Mater Res Part A. 108:2263–2276. 2020. View Article : Google Scholar | |
El-Housiny S, Eldeen MA, El-Attar YA, Salem HA, Attia D, Bendas ER and El-Nabarawi MA: Fluconazole-loaded solid lipid nanoparticles topical gel for treatment of pityriasis versicolor: Formulation and clinical study. Drug Deliv. 25:78–90. 2018. View Article : Google Scholar : PubMed/NCBI | |
Raad I, Mohamed JA, Reitzel RA, Jiang Y, Raad S, Al Shuaibi M, Chaftari AM and Hachem RY: Improved antibiotic-impregnated catheters with extended-spectrum activity against resistant bacteria and fungi. Antimicrob Agents Chemother. 56:935–941. 2012. View Article : Google Scholar : PubMed/NCBI |