Next Article in Journal
The CARD9 Gene in Koalas (Phascolarctos cinereus): Does It Play a Role in the Cryptococcus–Koala Interaction?
Previous Article in Journal
A Multiomics Perspective on Plant Cell Wall-Degrading Enzyme Production: Insights from the Unexploited Fungus Trichoderma erinaceum
Previous Article in Special Issue
Potential Activity of Micafungin and Amphotericin B Co-Encapsulated in Nanoemulsion against Systemic Candida auris Infection in a Mice Model
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Current Perspectives of Antifungal Therapy: A Special Focus on Candida auris

by
Arumugam Ganeshkumar
1,2,*,
Manickam Muthuselvam
3,
Patricia Michelle Nagai de Lima
1,
Rajendren Rajaram
4 and
Juliana Campos Junqueira
1,*
1
Department of Biosciences and Oral Diagnosis, Institute of Science and Technology, São Paulo State University (UNESP), São José dos Campos 12245-000, SP, Brazil
2
Department of Materials Physics, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences (SIMTS), Chennai 602105, Tamil Nadu, India
3
Department of Biotechnology, Bharathidasan University, Tiruchirappalli 620024, Tamil Nadu, India
4
Department of Marine Science, Bharathidasan University, Tiruchirappalli 620024, Tamil Nadu, India
*
Authors to whom correspondence should be addressed.
J. Fungi 2024, 10(6), 408; https://doi.org/10.3390/jof10060408
Submission received: 10 May 2024 / Revised: 29 May 2024 / Accepted: 3 June 2024 / Published: 6 June 2024
(This article belongs to the Special Issue Alternative Therapeutic Approaches of Candida Infections, 3rd Edition)

Abstract

:
Candida auris is an emerging Candida sp. that has rapidly spread all over the world. The evidence regarding its origin and emerging resistance is still unclear. The severe infection caused by this species results in significant mortality and morbidity among the elderly and immunocompromised individuals. The development of drug resistance is the major factor associated with the therapeutic failure of existing antifungal agents. Previous studies have addressed the antifungal resistance profile and drug discovery for C. auris. However, complete coverage of this information in a single investigation is not yet available. In this review, we have mainly focused on recent developments in therapeutic strategies against C. auris. Based on the available information, several different approaches were discussed, including existing antifungal drugs, chemical compounds, essential oils, natural products, antifungal peptides, immunotherapy, antimicrobial photodynamic therapy, drug repurposing, and drug delivery systems. Among them, synthetic chemicals, natural products, and antifungal peptides are the prime contributors. However, a limited number of resources are available to prove the efficiency of these potential therapies in clinical usage. Therefore, we anticipate that the findings gathered in this review will encourage further in vivo studies and clinical trials.

1. Introduction

1.1. Candidiasis, Outbreak, and Epidemiology

Candidiasis is an infection caused by opportunistic pathogens of the Candida genus, with manifestations varying from mucocutaneous lesions to life threading bloodstream infections [1,2]. Candida albicans is the most common Candida species found in various human anatomical sites, including oropharyngeal, esophageal, gastrointestinal, and genital mucosa [3]. Other non-albicans species and Candida-related species are also simultaneously reported in the human body, such as Nakaseomyces glabratus (formerly Candida glabrata), Pichia kudriavzevii (formerly Candida krusei), Candida parapsilosis, Candida tropicalis, and others [1,4]. Recently, an emerging pathogen named Candida auris has been isolated from various clinical samples such as urine, stool, and vaginal, and rectal swabs. Similar to other Candida species, patients with comorbidities or a weakened immune system, or who have previously been exposed to antifungals or subjected to prolonged stays in healthcare settings are at risk for C. auris infection [5,6].
C. auris is recognized as an emerging fungal pathogen because of its widespread distribution, multidrug-resistant (MDR) behavior, high transmissibility, strong association with nosocomial infections and high mortality rates. C. auris expresses numerous virulence traits as well as tolerance to common antifungals, leading to therapeutic failure when treated with the most common classes of antifungals, including azoles, polyene, and echinocandins. In 2009, the first isolate of C. auris was reported from a Japanese female patient with ear discharge. In the same year, twelve isolates were obtained from otitis media patients in South Korea. Between 2009 and 2011, twelve isolates of C. auris were identified in India in patients with bloodstream infections. The first outbreak was noted during 2016 and 2017 in Europe and the United States of America (USA), respectively. Subsequently, C. auris has been reported in more than 47 countries according to the Centre for Disease Control (CDC) [7,8], and, nowadays, it is recognized as a threat to global society by the World Health Organization (WHO). Based on susceptibility profiles, outbreak potential, and clinical manifestations, C. auris isolates have been broadly classified into five clades (Figure 1). Recently, unpublished data indicated a sixth clade of C. auris isolated from samples collected in Singapore. The uncommon Clade VI isolates differed significantly from all other isolates in terms of their antifungal resistance genes, mating type locus, and chromosomal rearrangements. Although C. auris isolates have been widely investigated, the real rate of prevalence remains uncertain due to the lack of a proper dataset [9].
Furthermore, the origin of C. auris has not yet been elucidated and some authors speculate that global warming may be a possible reason for its spread [10]. C. auris cells remain viable for several months on environmental surfaces and medical equipment and prefer to colonize the skin of patients rather than other mucosal surfaces, leading to high probability of person-to-person transmission [11]. Since cells live freely on biotic and abiotic surfaces and tolerate antifungal and decontamination agents, the eradication of these fungal cells is extremely complicated [12,13,14,15]. All these factors make C. auris a global threat for immunocompromised patients in healthcare settings.

1.2. Drug Resistance: Molecular Bases

Clinical breakpoints of the existing antifungals for C. auris still need to be established. However, CDC has recommended the use of clinical breakpoints established from other closely related Candida species. The general guidelines defined to correlate the antifungal resistance of C. auris are ≥32, ≥2, ≥4, ≥2, and ≥4 for fluconazole, amphotericin B (AmB), anidulafungin, caspofungin, and micafungin, respectively [16]. Most clinical isolates of C. auris are multidrug-resistant or pan-resistant, especially for fluconazole, followed by AmB and voriconazole [17]. The wide administration of fluconazole for Candida prophylaxis and the use of azole derivatives in various applications, such as agriculture, might be the primary factors contributing to the development of azole resistance. Echinocandins are the only choice for C. auris infection treatment, although some studies already reported echinocandin-resistant strains [18].
The prevalence of drug-resistant C. auris strains has been analyzed in several studies worldwide. Isolates from India, Pakistan, South Africa, and Venezuela presented resistance rates of 93% to fluconazole, 35% to amphotericin B (AmB), and 7% to echinocandins, respectively. [6]. A resistance rate of 90% for fluconazole was observed in isolates from India and the USA, whereas all the isolates from the United Kingdom (UK) showed 100% resistance to fluconazole. AmB was the second most common antifungal drug related to C. auris resistance, with 20–30% of resistant isolates in India [19], 30% in USA [20], 62.5% in Saudi Arabia [21], 23.2% in Kuwait [22], and 33.3% in Oman [23].
Recent studies investigated the molecular bases of drug resistance in C. auris isolates. Different mechanisms of azole resistance have been discussed, including mutation in target genes (ERG11) [24], overexpression of genes encoding drug transporters, and an increased copy number of the TAC1B gene [25]. Mutation in the TAC1B gene was the second most observed mutation in C. auris after ERG11. Mutations like F126L, Y132F, and K143R were frequently noted in the ERG11 gene in fluconazole-resistant isolates, whereas A640V, A657V, and F862_N866del mutations were observed in the TAC1B gene in fluconazole-resistant isolates [25,26].
Mechanisms of resistance to echinocandins are mainly associated with the FKS1 gene involved in the biosynthesis of glucan and maintains the integrity of the cell wall. As occurs with C. albicans and C. glabrata, FKS1 is the target of echinocandins and mutations such as amino acid substitution and deletion in hot spots of FKS1 result in a lack of therapeutic effectiveness [27,28]. The most frequently observed FKS1 mutations are S639F, S639P, and S639Y in hot spot 1; however, less common mutations in hot spot 1 have also been reported (F635 del substitutions, F635L/Y, S639T, and D642Y) [29,30].
Regarding AmB resistance, a probable mechanism of action has not yet been elucidated. The increased Minimum Inhibitory Concentration (MIC) observed in AmB-resistant C. auris isolates were associated with mutation in the ERG6 gene [29]. Due to a loss of sterol methyltransferase activity, these isolates accumulate more cholesta-type sterols [26] Similarly, in vitro studies have evidenced that AmB-resistant strains carry nonsense mutations in genes like ERG11 and ERG3 [24].

2. Therapeutic Options

2.1. Current Approved Antifungal/Considered Antifungals

The antifungal pipeline for C. auris infections includes various promising antifungal candidates, such as rezafungin, ibrexafungerp (IBX), fosmanogepix, and T-2307 (Figure 2). Rezafungin is a potential antifungal derived from anidulafungin that has exhibited strong in vitro antifungal activity against C. auris, including strains resistant to other echinocandins. It features a long half-life allowing for a once-weekly dosing schedule. In clinical trials, rezafungin has demonstrated a favorable safety profile with minimal side effects. On 31 March 2023, Rezzayo™ (Cidara Therapeutics, Inc., San Diego, CA, USA) was approved in the USA to treat adults with candidemia and invasive candidiasis. Nevertheless, it is still crucial to confirm its efficiency through data obtained from other clinical investigations. The high cost of this antifungal puts a barrier to its use in clinical practice in comparison with conventional treatments.
Due to their notable superior antifungal activity, researchers have been focusing on IBX. This drug is a class of (1,3) β-D-glucan synthase inhibitors like echinocandins, a semi-synthetic derivate of enfumafungin. The mechanism of binding of IBX was not disturbed by the FKS1 gene mutation, which limits the cross resistance [32]. During preclinical investigations, its potential as a therapeutic agent for managing highly resistant Candida infections was confirmed [33,34]. IBX remains active against fluconazole and echinocandin-resistant Candida isolates and multidrug-resistant C. auris. However, IBX has shown a concentration-dependent fungicidal activity against various clinically relevant fungi, including C. auris [33,34,35,36].
Fosmanogepix, also known as APX001, is also a promising antifungal agent exhibiting the potential to treat C. auris infections. Fosmanogepix is a prodrug that is actively converted into manogepix, the active molecule. Manogepix directly inhibits the function of the Gwt1 (cell wall transfer protein 1) enzyme, resulting in pleiotropic effects on fungal cells, leading to severe growth defects and cell death. Clinical trials provided encouraging results regarding its efficacy and safety against this multidrug-resistant fungus. Fosmanogepix demonstrated strong activity against C. auris strains in laboratory tests, including those resistant to standard antifungals such as echinocandins. Phase II trials evaluating fosmanogepix against C. auris infections showed high treatment success rates. Apart from C. auris, fosmanogepix exhibited a broad spectrum of activity against other Candida species and filamentous fungus, making it a valuable tool for wider fungal infections. Clinical trials indicated that fosmanogepix was well-tolerated with minimal side effects. Serious adverse effects or treatment discontinuations were not reported. Fosmanogepix is available in both intravenous and oral forms, offering flexibility in treatment administration based on patient needs and disease severity [37]. Multiple Phase III clinical trials are currently investigating fosmanogepix for different fungal infections, including invasive candidiasis caused by C. auris. These trials will further evaluate its efficacy and safety on a larger patient population. Fosmanogepix is not yet approved for clinical use in any country. While the promising results from Phase II trials are encouraging, further research and evaluation through Phase III trials are still required [38].
Interestingly, a novel arylamidine (T-2307) expressed broad-spectrum antifungal activity against various fungal pathogens including several Candida species. Recent investigations highlighted the in vitro and in vivo efficiency of T-2307 on C. auris. Its mechanism of action was associated with the collapse of mitochondrial membrane potential by targeting respiratory chain enzymatic complexes III and IV of the yeast [39,40,41]. Phase I clinical trials evaluating the safety and tolerability of T-2307 in healthy volunteers were recently completed, showing promising results. Phase II clinical trials to assess its efficacy in patients with fungal infections are planned to begin in 2024. Considering its promising preclinical data and ongoing clinical trials, T-2307 could be approved for clinical use against C. auris infections within the next few years.

2.2. Chemicals as an Emerging Weapon against C. auris

Whilst some antifungal agents are in the clinical trials phase, the scientific community has worked intensely to discover other therapeutic options. Here, several new compounds with activity against C. auris are listed (Table 1) and discussed. Toepfer and collaborators investigated the compound clorgyline and its derivatives, which worked as multi-targeting inhibitors of the Cdr1 and Mdr1 efflux pumps of C. albicans and C. glabrata. Especially, the clorgyline analogs M19 and M25 expressed a high ability to inhibit the efflux pump activity of C. auris [42]. Recent investigations have discovered the activity of pyrazole moiety compounds against fluconazole-resistant C. auris isolates. These compounds exhibited broad spectrum, high potency, high selectivity, low cytotoxicity, and anti-drug resistance [43]. Novel tetrazoles featuring isoxazole moiety have also been identified as highly selective antifungal agents, displaying outstanding antifungal activity against fluconazole-resistant strains of C. albicans, C. glabrata, and C. auris [44]. A novel benzoanilide antifungal (compound A1) showed potent activity against C. auris cells through the blocking of virulence biosynthesis and altering of the cell wall of C. auris [45]. Furthermore, a compound named NSC319726 (thiosemicarbazone zinc chelator) exhibited MIC values ranging from 0.125 to 0.25 mg/L for C. auris isolates belonging to five different clades, expressing fungistatic activity in time-kill curves [46].
Many authors have focused on the study of Manogepix, a pyridine-isoxazole-based antifungal agent. Manogepix was active against most clinical isolates of C. auris belonging to South Africa. Over 300 C. auris isolates were studied, including 335 fluconazole-resistant, 19 fluconazole and AmB-resistant, 1 AmB-resistant and 2 pan-resistant. The MIC values of manogepix ranged from 0.002 to 0.063 μg/mL for fluconazole-resistant isolates, 0.004 to 0.031 μg/mL for fluconazole and AmB-resistant isolates, and 0.004 μg/mL to 0.008 μg/mL for pan-resistant isolates. The activity of manogepix was higher than azoles (3-fold), 4-fold higher than echinocandins, and 9-fold higher than AmB [50]. Manogepix also showed activity against clinical isolates from the New York outbreak, with MIC values of 0.008 to 0.015 mg/L against pan-resistant isolates [51]. A recent study has observed an increase in efficiency of anidulafungin against C. auris when combined with manogepix or 5-flucytosine [55].
Various studies have reported the biological role of metallic gold and its salts. Among them, gold(I)—phosphine complexes and the gold salt auranofin were tested against a panel of 28 fungal strains including Candida spp., Cryptococcus spp., Aspergillus spp., and Fusarium spp. Notably, two (complex 4 and 6) square-planar gold(I) complexes produced a remarkable antifungal activity in most of the tested isolates. Regarding C. auris isolates, complex 4 and 6 resulted in MIC values ranging from 3.9 to 7.8, and 1.95 µg/mL, respectively. However, auranofin did not produce considerable results (MIC > 31.3 µg/mL) against Candida species [52].
The activity of new promising antifungal compounds has also been investigated against C. auris in biofilm stage. Among several phenylthiazole small molecules, compound 1 emerged as the most potent antifungal, inhibiting the growth of C. albicans and C. auris strains at concentrations ranging from 0.25 to 2 µg/mL. This compound reduced 50% of metabolic activity of biofilm produced by C. auris with similar activity to AmB [53]. A class of molecule ceragenins was also found to inhibit both the planktonic and biofilm form of C. auris. Promisingly, they led to significant reductions in fungal infections in ex vivo mucosal tissues [54].

2.3. Essential Oils Are Potential Sources of Novel Antifungal Skeletons

Essential oils are mixtures of volatile compounds obtained through the steam distillation of various plant parts, such as flowers, bark, leaves, and other plant materials. Due to their unique properties, essential oils have a wide range of applications, with a predominant use in aromatherapy. Due to the presence of diversified volatile compounds, essential oils have been extensively studied as antimicrobial agents against bacteria and fungi, including C. auris strains.
For instance, Parker et al. demonstrated the effectiveness of selected essential oils against C. auris and found superior activity for the oils from cinnamon leaf, clove bud, lemongrass, and basil. The effective eradication of C. auris occurred with MIC values ranged from 0.01% to 1.0%. The same study reported the interactions between conventional antifungal drugs and essential oils. Clove bud oil synergistically interacted with fluconazole and flucytosine to combat C. auris [56].
Essential oil extracted from the seeds of Withania somnifera was also tested against C. auris, producing IC50 at 5.96 mg/mL and a fungistatic mechanism confirmed by killing assay. Its mechanism of action was associated with disturbing the membrane integrity of C. auris cells, evidenced through ergosterol binding and sorbitol protection assays. However, the seed oil was inactive against the mature biofilm formed by C. auris [57].
In a more detailed study, Di Vito et al. tested 15 essential oils against 10 clinical strains of C. auris. The results indicated that Cinnamomum zeylanicum essential oil was most effective against C. auris (MIC; 0.06% v/v) in synergy with the antifungal drug fluconazole. Further they verified that cinnamaldehyde was the sole reason for the antifungal activity [58]. In the same year, another group of researchers identified the antifungal potency of Cinnamomum cassia essential oil [59], this oil also being rich in chemical compounds like cinnamaldehyde [60]. The level of cinnamaldehyde in plants may vary depending upon the species; however, C. cassia and C. zeylanicum contain 85.3% and 90.5% cinnamaldehyde respectively [61].

2.4. Natural Products against Candida auris

The world of medicine is increasingly turning to nature’s bounty for solutions to modern health challenges. In the realm of fungal infections, a fascinating arsenal of weapons lies hidden within plants, microbes, and even marine organisms. These diverse and potent molecules, meticulously crafted by living organisms, offer a promising alternative to traditional antifungal drugs. With the rising tide of fungal resistance to existing therapies, natural products present a glimmer of hope in the fight against these tenacious pathogens. A list of natural products with anticandidal activity is presented in Table 2 and Figure 3.
Among the compounds from plants, great focus has been given to carvacrol, a phenolic monoterpenoid found in the essential oils of oregano, thyme, pepperwort, wild bergamot, and other plants. Due to its broad spectrum of biological responses [73]. Carvacrol was active against C. auris by modulating the expression level and action of certain antioxidant enzymes [69]. As with carvacrol, geraniol is another monoterpene alcohol found in geranium oil as a major component. Recently, Fatima and collaborators utilized geraniol against C. auris. Geraniol displayed fungicidal activity and an inhibition effect on the metabolically active biofilm of C. auris. Subsequently, geraniol improved the survival rate of C. elegans infected by C. auris [70].
Penta-O-galloyl-β-D-glucose (PGG), a bioactive product of many plants (firstly isolated from the leaves of Schinus terebinthifolia), was also investigated against C. auris. Chemically, PGG is a hydrolysable tannin reported to have plenty of biological activities such as antibacterial, anticancer, and antiviral activities. PGG demonstrated anticandidal activity within the MIC ranges of 1–8 µg/mL against drug-resistant C. auris [63]. Kim and Eom explored the antifungal and anti-biofilm properties of 6-shogaol against C. auris. Shogaols are pungent constituents of ginger and similar in chemical structure to gingerol [74]. The 6-shogaol demonstrated effectiveness in inhibiting the growth of C. auris in the concentration range of 16–32 µg/mL. Further, it showed promising activity in preventing the formation of biofilms and controlled the secreted aspartyl proteinase activity [72].
In relation to natural compounds from microbial sources, rubiginosin C obtained from the stromata of the ascomycetes Hypoxylon rubiginosum and Hypoxylon texense, effectively inhibited the formation of biofilms of C. auris and C. albicans [75]. In recent years, many studies have focused on the compound enfumafungin, a triterpene glycoside found in the culture supernatant of Hormonema carpetanum. This compound was used as a probe to produce biologically active antifungal IBX. Enfumafungin B and C, analogues of enfumafungin, were isolated. Both compounds were effective against clinically relevant C. auris with a MIC of 64 µg/mL. Further molecular docking studies confirmed that these compounds bonded in the transmembrane region of FKS1 of β-(1,3)-D-glucansynthase [64].
In another study, Persephacin, isolated from the endophytic fungus Sphaceloma sp., showed activity against a diverse range of fungal species (C. albicans, C. glabrata, C. parapsilosis, C. krusei, C. kefyr, C. tropicalis, and C. auris). Persephacin resulted in MIC values around 2.5 µg/mL, which was similar to the activity expressed by AmB [65]. A group of new linear lipopeptides (Myropeptin C–E and Myropeptin A1), isolated from the saprotrophic filamentous fungus Myrothecium inundatum, showed good inhibition of C. auris cells. In vitro hemolysis, cell viability, and ionophore assays indicated that these compounds target mitochondrial and cellular membranes, inducing cell depolarization and cell death [66]. Subsequently, hakuhybotrol along with six known cladobotric acids F, E, H, A, pyrenulic acid A, and F2928-1 were isolated from a culture broth of Hypomyces pseudocorticiicola FKA-73. Most of these compounds showed promising antifungal activity against azole-resistant and sensitive strains of C. auris. In particular, cladobotric acids F and E were able to cause a high inhibition in the fungal growth [67].
Although most antifungal compounds from microbial sources were isolated from fungus, some studies have investigated compounds from bacterial cultures. For example, a culture extract of Lactobacillus paracasei 28.4 inhibited several C. auris strains, acting against planktonic cells, biofilms, and persister cells. Further experiments confirmed that supplementation derived from L. paracasei 28.4 protected G. mellonella from C. auris infection [68].
Finally, compounds from marine sources have also been investigated against C. auris. Turbinmicin, a potent lead obtained from the marine reservoir (Turbinmicin-producing bacterium Micromonospora sp. WMMC-415.), displayed antifungal properties in several in vitro and in vivo experiments. Turbinmicin expressed a fungal-specific mechanism of action, targeting Sec14 of the vesicular trafficking pathway, a unique target, yet to be investigated [62]. A subsequent investigation showed that Turbinmicin exhibited a MIC value of 0.125 mg/mL and an inhibitory action on the mature biofilm of C. auris [76].

2.5. Peptide-Based Strategies for Eradicating C. auris

Antimicrobial peptides (AMPs) are another alternative group of components frequently reported due to their superior biological properties. They are an active form of smaller segments of protein produced by various organisms, including plants, insects, humans, and other small animals. AMPs have distinct functions within the host; therefore, they express unexceptional behavior towards medically important pathogens like bacteria, fungi, and virus. On the other hand, the antimicrobial resistance mechanism of AMPs has not yet been fully elucidated. Wider investigations have identified some direct or indirect mechanisms of AMPs against pathogens, with a capacity to reduce virulence traits. As per the information available [77], there have been more than 3940 AMPs reported until now, including 3146 natural peptides and 190 predicted and 314 synthetic AMPs. More specifically, HsAFP1 (Heuchera sanguinea); NaD1 (Nicotiana alata flowers); Psd1 (Pisum sativum seeds); Psoriasin, CGA-N46, β-Defensin-1 to 4, and Histatin-5 from Homo sapiens; Gomesin, Heliomicin, Jelleine I to IV, and Lasioglossin I to III from insects and arachnids; and NFAP2 from filamentous fungi Neosartorya fischeri have been reported to have anticandidal activity [78]. A list of AMPs with their significant biological roles and their origins is presented in Table 3.
Among the microorganisms, both fungi and bacteria produce potent antimicrobial peptides. The fungus Neosartorya fischeri produces two distinct peptides: NFAP and NFAP2, with 57 and 52 amino acid lengths, respectively. The cystine residues present in these structures have remarkable importance, since they significantly improve the stability of the peptides at high temperatures [79,80]. NFAP and NFAP2 were identified as potent molecules against fluconazole (FLC)-resistant C. albicans [81] and C. auris [82]. Notoriously, NFAP2 interacted with most of the azoles and echinocandins, producing significant Fractional Inhibitory Concentration Index (FICI) values [82]. The bacteria Bacillus subtilis produces a lipopeptide (AF4) that showed a broad spectrum of antifungal activity on more than 110 fungal isolates. Recently, AF4 at 8 mg/L was found to kill most of C. auris cells. The mechanism of killing was associated with severe cellular membrane disruption and elevated generation of Reactive Oxygen Species (ROS) [83].
Table 3. Antifungal peptides against C. auris derived from different classes of organisms.
Table 3. Antifungal peptides against C. auris derived from different classes of organisms.
S.noOriginName MIC Range Reference
1Homo sapiensHuman β-defensin-3 3.125 to 12.5 µg/mLShaban et al., 2023 [84]
2Homo sapiensCathelicidin peptides LL-3725–100 µg/mLRather et al., 2022 [85]
3Homo sapiensHistatin-5 7.5 µMPathirana et al., 2018 [86]
4Barley plantDefensin-like Protein 1 (D-lp1)0.047–0.78 mg/mLKamli et al., 2022 [87]
5Bacillus subtilisAF4 8 µg/mLRamesh et al., 2023 [83]
6American rattlesnake (Crotalus durissus terrificus)Crotamine 40–80 µMDal Mas et al., 2019 [88]
7Pomacea poeyana—freshwater snailPom-1, Pom-28.5, 8.4 µg/mLRaber et al., 2021 [89]
8Scorpion venomToAP1, ToAP2>100 µM, 50–>100 µMPinheiro et al., 2023 [90]
9BrilacidinSemi synthetic80 µg/mLDos Reis et al., 2023 [91]
10Chemically prepared, Myristoylated and non-Myristoylated peptidesPep-A, Myr-A, Pep-B, Myr-B, Pep-C, Myr-C>256, >256, >256, 16–32, >256, 16–64Bugli et al., 2022 [92]
11Symbiotic NCR peptide fragmentsNCR169C 17–386.25 µMSzerencsés et al., 2021 [93]
NCR169C 17–38 ox12.5 µM
12Analogue of the peptide Cm-p5
MonomersCm-p5, Cyclic, Hcy11 µg/mL, 27 µg/mL, Not active Vicente et al., 2019 [94]
Dimer Dimer 1 (parallel)30 µg/mL
Dimer 2 (anti-parallel)31 µg/mL
13Rhesus macaque θ-defensin (RTD)RTD–1, RTD–26.25, 6.25 µg/mLBasso et al., 2018 [95]
Olive baboon θ-defensins (BTD)BTD–2, BTD–4, BTD–83.125, >25, 3.12–6.25 µg/mL
The human body is also a significant source of antifungal peptides. So far, many peptides with different biological functions have been reported from the human body. Peptides like human β-defensin-3 [84], human cathelicidin peptides LL-37 [85], and salivary histatin-5 [86] were recently reported to have antifungal activity on C. auris (Table 3). Human β-defensin-3 and cathelicidin peptides LL-37 produced 100% and 70% of synergy with fluconazole [84,85]. The anticandidal activity of human histatin-5 was also documented against other non-albicans species, including C. glabrata, C. parapsilosis, C. krusei, C. guilliermondii, and C. tropicalis [96]. In addition, some studies highlighted the ex vivo effects of histatin-5 on mouse models, by reducing the fungal burden in both oral and vaginal infection models [97,98].
Plant defensins are potent molecules that protect plants from infections by effectively combating microbes without harming host cells. Certain plant-derived peptides, like HsAFP1 [99], NaD1 [100], Psd1 [101], and D-lp1 [102] exhibited strong activity against C. auris. Among them, D-lp1completely inhibited the biofilm formation and virulence of C. auris [102]. Similar to plant AMPs, other living organisms were found to be a source of antifungal molecules. Crotamine (from a South American rattlesnake) acted on multidrug-resistant C. auris with no cytotoxicity [88]. Pom-1 and Pom-2 peptides from Pomacea poeyana (the Cuban freshwater snail) inhibited Pseudomonas aeruginosa and fungi, including C. auris [89,102]. Scorpion venom peptides ToAP1 and ToAP2 showed promising antifungal effects against C. auris, alone and associated with other antimicrobial drugs [90] (Table 3).

2.6. Antifungal Immune Therapy against C. auris

Apart from AMPs, there are some other proteinous molecules that arouse interest as therapeutic strategies towards C. auris due to their immunological properties. These groups of molecules can be derived from the immune system of humans or certain animals. Amongst them, the complement receptor 3-related protein (CR3-RP) is one of the key surface antigens expressed during the biofilm formation of Candida species. Previous investigation identified the presence of CR3-RP moieties on the surface of C. auris. Upon in vitro exposure to prepared anti-CR3-RP, C. auris cells failed to form biofilm, confirming the ability of anti-CR3-RP for eradicating C. auris biofilms [103]. Similarly, Singh et al. utilized the anti-Hyr1p monoclonal antibody (mAb) to control the C. auris infection. The anti-Hyr1p mAb prevented biofilm formation and enhanced opsonophagocytic killing of C. auris by macrophages. In vivo studies showed that anti-Hyr1p mAb protected 55% of mice from the systemic infection caused by C. auris [104]. Other than these, NDV-3A (a vaccine based on the N-terminus of the Als3 protein formulated with alum) also showed effects against C. auris, inhibiting biofilm formation and encouraging the macrophage-mediated killing of C. auris [105].
A new humanized antibody H5K1 was recently identified and found to be active against C. auris. H5K1 expressed significant results when tested alone or in combination with Caspofungin and AmB [106]. Recent findings suggested that H5K1 specifically binds to β-1,3-glucans derived from C. auris, causing perturbation and remodeling of the fungal cell wall and facilitating the loss of cellular membrane integrity [107]. In support of this investigation, other Candida cell surface-specific mAbs were investigated in a mouse model of a C. auris invasive infection. For example, the specific monoclonal antibody C3.1, which targets the β-1,2-mannotriose (β-Man3) of C. auris, was able to improve the survival of animals and reduce the fungal burden in vital organs. In the same study, other peptide-specific mAbs such as 6H1 and 9F2 were reported as targeting hyphal-specific protein 1 (Hwp1) and phosphoglycerate kinase 1 (Pgk1), respectively. It also showed the same outcome as C3.1 in comparison with the control group. Altogether, the 6H1 + 9F2 cocktail enhanced the therapeutic outcome compared to the monotherapy. Therefore, all the three antibodies reported here might be an alternative to treat C. auris infections [108].
Intravenous immunoglobulins (IVIG) have been considered as an alternative therapeutic strategy to treat patients who present primary antibody deficiencies. They are the therapeutic product of normal human IgG [109,110,111]. Xin et al. recently demonstrated the role of IVIG in prevention and control of C. auris and C. albicans invasive candidiasis in animal models. Treatment with IVIG prolonged the survival and reduced the fungal burden in organs of the treated animals. In combination, IVIG enhanced the therapeutic index of AmB in comparison with monotherapy [112].
Immuno-informatics-based approaches offer an alternative and burgeoning avenue for designing suitable vaccine candidates against fungal infections. Recently, subtractive proteomics approaches were employed to design vaccines against C. auris. Khan et al. adopted this method and generated multi-epitope vaccine candidates, which elicited immune responses against C. auris infection by inducing various immune factors such as IgM, IgG, IL-6, and Interferon-α [113]. Similarly, Gupta and colleagues developed a vaccine based on novel CD4+ epitopes through genome-wide scanning and a reverse vaccinology approach. This vaccine has a reduced chance of becoming ineffective because the rapidly evolving genes of C. auris were eliminated from the epitope selection process [114].

2.7. Antimicrobial Photodynamic Therapy (APDT) against C. auris

APDT is a promising approach as an adjuvant therapy for fungal infections. As an advantage, APDT simultaneously targets different biomolecules of pathogens. Thus, it does not have any specific mechanism of action, which restricts the development of cross resistance. Therefore, APDT is considered an alternative therapeutic strategy against multiple clinically important organisms, including C. auris. APDT involves the association of a photosensitizer with irradiation from a light source, resulting in the generation of ROS, a prime factor responsible for the effectiveness of APDT.
Phenothiazinium-based photosensitizers are promising agents proven to deactivate C. auris cells. Methylene blue, toluidine blue, new methylene blue, and the pentacyclic derivative S137 were assessed as photosensitizers for APDT on C. auris (CDC B11903). Their efficacy was evaluated by the determination of MIC and the use of a G. mellonella insect model. Based on the findings, the pentacyclic derivative S137 was identified as a potent treatment for C. auris [115]. Previously, it had demonstrated strong inhibition of C. albicans [116]. In another study, researchers utilized red, green, and blue visible lights alone and in combination with photosensitizers (new methylene blue, toluidine blue O, and rose bengal) against C. auris. The results showed that blue light alone disturbed the mature biofilm, but it was significantly improved when the photosensitizer was combined. On the other hand, red or green light alone had no effect on the Candida biofilm. The biofilms were disturbed only with the combination of light and photosensitizers [117].
Recently, Silva and their coworkers assessed the impact of methylene blue and 1,9-dimethyl methylene blue in addition with a red light-emitting diode (LED) on C. auris. At 3 μM, regardless of the light dose, 1,9-dimethyl methylene blue reduced the metabolic activity of Candida cells. Furthermore, it promoted high levels of ROS, lipid peroxidation, and mitochondrial membrane damage. In contrast, methylene blue was active only at a concentration of 100 μM when exposed to the highest dose of light. Further, studies evidenced that 1,9-dimethyl methylene blue was capable of inhibiting biofilm formation and the mature biofilm formed by C. auris [118]. Earlier studies by Stefanek and his team confirmed the positive effect of methylene blue with a red laser on the biofilm of C. auris. Up to 90% biofilm inhibition after 300 s of irradiation was observed compared to the growth control. Interestingly, in the presence of 0.25 mM methylene blue, the expression of both MDR1 and CDR1 genes was affected [119].

2.8. Repurposing of Drugs with Antifungal Properties

Drug repositioning or repurposing is a process of utilizing commercially available drugs for treating diseases outside the scope of its original indication. Drug repurposing is considered an important approach to manage emerging diseases caused by bacteria, fungi, and viruses [120,121]. The availability of various information rather than therapeutic indexes is a valuable point to consider in the repurposing process. Drug repurposing can reduce the time and cost of new drug development since it leverages existing data on toxicity profiles and preclinical parameters [122]. Therefore, the repurposing of drugs is recognized as an alternative to combat antifungal drug resistance, and several commercial non-antifungal drugs with activity against C. auris have been reported (Figure 4). Among them, Sertraline (an antidepressant agent that comes under serotonin reuptake inhibitors) showed activity against three different isolates of C. auris, possessing efficient antifungal activity by suppressing the action of yeast to hyphae conversion and biofilm formation [123].
Lohse and their colleague have aimed to develop antifungal agents from a group of FDA-approved compounds [124]. The selection of these compounds was based on MIC values of <10 µM. Among the hydroxyquinolines tested, clioquinol was more active than others; however, the authors were unable to investigate their mechanisms of action [124]. In another study, a hydroxyquinoline derivative known as nitroxoline was also active against 35 isolates of C. auris with MIC values ranging from 0.125 to 1 μg/mL. The resultant MIC values were lesser than the activity of fluconazole and AmB. Finally, nitroxoline was recommended for the treatment of C. auris-mediated candiduria. However, in vivo and clinical efficiency remains uncertain [49].
Synergistic drug interactions have also been investigated to increase the success of drug repurposing [125]. Pitavastatin, a cholesterol-lowering drug, proved to be a potent azole chemosensitizer. It reduced Candida biofilm formation and lowered MIC ranges when combined with fluconazole against C. auris [126]. Aprepitant, an antiemetic drug, showed the ability to disrupt metal ion homeostasis in C. auris, synergizing with azoles to reduce MIC by up to eight-fold and inhibiting biofilm formation by 95 ± 0.13% [127]. Miltefosine, an antiparasitic drug licensed for leishmaniasis, demonstrated potential against C. auris and other Candida strains, especially in combination with other antifungal drugs [128,129]. Colistin, an antibiotic used against multidrug-resistant Gram-negative infections, such as pneumonia, showed synergistic effects when combined with caspofungin, with FICI values ranging from 0.08 to 0.14. However, combining colistin with micafungin showed indifferent results, with FICI values ranging from 0.51 to 1.01 [130]. Synergistic combinations between HIV protease inhibitors and azoles were also found to be active against drug-resistant C. auris. Lopinavir combined with itraconazole achieved potent effects, increasing the survival rate of C. auris-infected C. elegans by up to 90% and reducing fungal burden by 88.5% [131]. Additionally, lopinavir and ritonavir interacted synergistically with itraconazole, effectively combating disseminated candidiasis in a rat model [132]. Atazanavir resensitizes C. auris to azoles by inhibiting efflux pumps, glucose transport, and ATP synthesis [133]. Moreover, the combination of saquinavir and itraconazole significantly reduced fungal burden in murine models, with an 88% decrease in colony-forming units compared to itraconazole alone [134].
Recently, some studies validated the synergistic potential of azoles in combination with chlorhexidine, used as a skin antiseptic and mouthwash due to its broad-spectrum antibacterial effects. It was reported that chlorhexidine can bind to cellular membrane phospholipids, causing changes in osmotic pressure and promoting cell lysis [135]. When combined with fluconazole, chlorhexidine significantly reduced the viability of both planktonic and biofilm forms of C. auris [136]. These results suggest the combined use of chlorhexidine and azoles to control C. auris infections in cutaneous and mucosal surfaces. Sulfamethoxazole is another class of antibiotic usually used to treat a variety of infections of the urinary tract, respiratory system, and gastrointestinal tract. Earlier investigation identified synergist interaction of sulfamethoxazole with azole antifungal drugs against C. albicans. The present study highlighted that sulfamethoxazole in combination with voriconazole and itraconazole restored the fungistatic potency of both drugs. Further, the superiority of the azole–sulfamethoxazole combination against C. auris infection was proved in C. elegans [137].

2.9. Nanotechnology-Mediated Antifungal Therapy

Metallic nanoparticles have been investigated as antimicrobial agents against a large number of microorganisms. The detailed investigations of the potent antifungal properties of different nanoparticles are presented in this review. In the last decades, researchers have extensively worked to develop different metallic nanoparticles, such as Ag, Zn, and Au, targeted to combat pathogenic microorganisms.
Regarding C. auris, most studies have focused on silver-based nanoparticles. Humberto and colleagues verified that silver nanoparticles effectively limited biofilm development at a silver nanoparticle concentration of 0.48 ppm, suggesting their use for controlling C. auris in healthcare settings [138]. Another study investigated silver nanoparticles for effectiveness against multidrug-resistant C. auris, showing strong antifungal properties with a MIC of <0.5 μg/mL on planktonic cells and a Minimum Biofilm Inhibitory Concentration (MBIC) of <2 μg/mL on preformed biofilm [139]. Consistent findings also revealed a significant reduction in viable C. auris cells, in both planktonic and biofilm form, upon treatment with silver nanoparticles [140].
More recently, several functionalized silver nanoparticles have been produced by green synthesis using plant compounds as metal ion-reducing agents. Polyphenol-capped metallic silver nanoparticles, such as those derived from Cynara cardunculus extract, exhibited an antifungal effect on C. auris by inducing mitochondrial toxicity and DNA fragmentation at a concentration of 50 µg/mL [141]. Trimetallic (Ag-Cu-Co) nanoparticles, synthesized with compounds from Salvia officinalis, also showed potent antifungal properties, inducing apoptosis and G2/M phase cell cycle arrest in C. auris, with MIC values ranging from 0.39 to 0.78 μg/mL and Minimum Fungicidal Concentrations (MFC) ranging from 0.78 to 1.56 μg/mL [142].
In addition to silver nanoparticles, various other metal nanoparticles have demonstrated activity against C. auris. For example, bismuth nanoparticles have shown promising effects in combating multidrug-resistant C. auris, exhibiting anticandidal activity with MIC ranging from 1 to 4 µg/mL, and disrupting both cells and biofilms of C. auris [143]. Caspofungin-loaded zinc oxide nanoparticles have demonstrated antifungal activity against caspofungin-resistant C. auris. Interestingly, caspofungin–ZnO nanoparticles did not induce acquired or cross resistance in C. auris [144].

2.10. Liposomal Technology for Efficient Drug Delivery

Liposomal technology represents a promising avenue for antifungal therapy, using various approaches to prepare liposomal delivery systems. The key point among these methods is the careful selection of lipid moieties for encapsulating a specific drug. This selection influences the surface charge of the liposome, enabling tailored delivery of potent molecules. The advantages of liposomal technology are manifold, including improved bioavailability, reduced toxicity, and targeted delivery. For instance, liposomal formulations of amphotericin B offer enhanced solubility and reduced nephrotoxicity compared to conventional amphotericin B, confirming the potential of liposomal technology in optimizing therapeutic outcomes.
De Alteriis et al. examined the role of liposomal technology to improve the antifungal effects of essential oil from Lavandula angustifolia, achieving antibiofilm activity with the persister-derived biofilm of C. auris [145]. Similarly, Lippia sidoides essential oil was loaded in lipid carriers, and the reduction in the MIC values were observed on C. auris [146]. In line with earlier investigations, Jaromin et al. utilized liposomal formulation to improve and modulate the surface characteristics of PQA-Az-13, which is the combination of indazole, pyrrolidine, and arylpiperazine scaffolds substituted with a trifluoromethyl moiety. Here, liposomes displayed a mean size of 76.4 nm, a positive charge of +45.0 mV, excellent stability, and no toxicity to normal human dermal fibroblasts. PQA-Az-13 showed MIC values between 0.67 and 1.25 µg/mL against C. auris and demonstrated promising results against in vitro biofilms and ex vivo skin colonization models [147].

3. Conclusions and Future Perspectives

The world is facing a crisis with the growing incidence of several infectious diseases and the limited availability of antifungal therapies, highlighting the urgent need to combat fungal infections. Compared to bacterial infections, fungal infections are less common, which has led to a limited range of antifungal drugs. However, targeting fungal-specific pathways plays a crucial role in the development of novel antifungal drugs. This limited choice of antifungal drugs contributes to their overuse, misuse, abuse, or prolonged use, leading to the development of antifungal drug resistance. The failure of antifungal therapy is associated with multiple factors, among which the development of drug resistance is the prime contributor. The identification of fungal-specific drug targets is a key characteristic of developing new antifungal therapies. This is crucial because they can effectively combat fungal infections without negatively affecting the host.
To address these challenges, there is an urgent need for innovative strategies in antifungal drug development. Especially, the elucidation of new drug targets, identification of novel drug scaffolds, designing of new drugs, and development of combined therapies are the most important points to consider. Additionally, some researchers are focusing on drug repurposing strategies. Adapting drugs already approved for other conditions can be a faster and cheaper way to bring new antifungals to market, especially against emerging or resistant fungal threats. While still in early stages, research on fungal vaccines is ongoing, focusing on stimulating the immune system to recognize and fight fungal infections. Tailoring antifungal treatment based on individual patient characteristics and the specific fungal strain involved in the infection can improve outcomes and reduce the risk of resistance.
Implementation of antimicrobial stewardship programs is crucial to prevent the overuse of common antifungal agents in medicine and other fields such as agriculture. By addressing these issues, there is a possibility to improve the management of severe fungal infections and reduce their impact on global health. Although several approaches discussed here showed promising antifungal activity against C. auris, few researchers extended their results to animal models and clinical trials. Overall, it is hoped that the data gathered in this review can provide support and insights into the advances of new treatments for C. auris infections.

Funding

This present work was carried out with the support of the Coordination for the Improvement of Higher Education Personnel—Brazil (CAPES)—88887.890668/2023-00. We are grateful to the Brazilian National Council for Scientific and Technological Development (CNPq 310265/2022-3) and office of Naval Research Global (N62909-20-1-2034).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data is contained within the article.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Pappas, P.G.; Lionakis, M.S.; Arendrup, M.C.; Ostrosky-Zeichner, L.; Kullberg, B.J. Invasive Candidiasis. Nat. Rev. Dis. Primers 2018, 4, 18026. [Google Scholar] [CrossRef]
  2. Tamo, S.P.B. Candida Infections: Clinical FeatuRes. Diagnosis and Treatment. Infect. Dis. Clin. Microbiol. 2020, 2, 91–102. [Google Scholar] [CrossRef]
  3. Talapko, J.; Juzbašić, M.; Matijević, T.; Pustijanac, E.; Bekić, S.; Kotris, I.; Škrlec, I. Candida albicans—The Virulence Factors and Clinical Manifestations of Infection. J. Fungi 2021, 7, 79. [Google Scholar] [CrossRef] [PubMed]
  4. Lamoth, F.; Lockhart, S.R.; Berkow, E.L.; Calandra, T. Changes in the Epidemiological Landscape of Invasive Candidiasis. J. Antimicrob. Chemother. 2018, 73 (Suppl. S1), i4–i13. [Google Scholar] [CrossRef]
  5. Frías-De-león, M.G.; Hernández-Castro, R.; Vite-Garín, T.; Arenas, R.; Bonifaz, A.; Castañón-OlivaRes, L.; Acosta-Altamirano, G.; Martínez-Herrera, E. Antifungal Resistance in Candida auris: Molecular Determinants. Antibiotics 2020, 9, 568. [Google Scholar] [CrossRef] [PubMed]
  6. Lockhart, S.R.; Etienne, K.A.; Vallabhaneni, S.; Farooqi, J.; Chowdhary, A.; Govender, N.P.; Colombo, A.L.; Calvo, B.; Cuomo, C.A.; Desjardins, C.A.; et al. Simultaneous Emergence of Multidrug-Resistant Candida auris on 3 Continents Confirmed by Whole-Genome Sequencing and Epidemiological Analyses. Clin. Infect Dis. 2017, 64, 134–140. [Google Scholar] [CrossRef] [PubMed]
  7. Mishra, S.K.; Yasir, M.; Willcox, M. Candida auris: An Emerging Antimicrobial-Resistant Organism with the Highest Level of Concern. Lancet Microbe 2023, 4, e482–e483. [Google Scholar] [CrossRef]
  8. Sanyaolu, A.; Okorie, C.; Marinkovic, A.; Abbasi, A.F.; Prakash, S.; Mangat, J.; Hosein, Z.; Haider, N.; Chan, J. Candida auris: An Overview of the Emerging Drug-Resistant Fungal Infection. Infect Chemother. 2022, 54, 236–246. [Google Scholar] [CrossRef]
  9. Suphavilai, C.; Ko, K.K.K.; Lim, K.M.; Tan, M.G.; Boonsimma, P.; Chu, J.J.K.; Goh, S.S.; Rajandran, P.; Lee, L.C.; Tan, K.Y.; et al. Discovery of the Sixth Candida auris Clade in Singapore. medRxiv 2023. [Google Scholar] [CrossRef]
  10. Adams, E.; Quinn, M.; Tsay, S.; Poirot, E.; Chaturvedi, S.; Southwick, K.; Greenko, J.; Fernandez, R.; Kallen, A.; Vallabhaneni, S.; et al. Candida auris in Healthcare Facilities, New York, USA, 2013–2017. Emerg. Infect. Dis. 2018, 24, 1816–1824. [Google Scholar] [CrossRef]
  11. Abdolrasouli, A.; Armstrong-James, D.; Ryan, L.; Schelenz, S. In Vitro Efficacy of Disinfectants Utilised for Skin Decolonisation and Environmental Decontamination during a Hospital Outbreak with Candida auris. Mycoses 2017, 60, 758–763. [Google Scholar] [CrossRef] [PubMed]
  12. Welsh, R.M.; Bentz, M.L.; Shams, A.; Houston, H.; Lyons, A.; Rose, L.J.; Litvintseva, A.P. Survival, Persistence, and Isolation of the Emerging Multidrug-Resistant Pathogenic Yeast Candida auris on a Plastic Health Care Surface. J. Clin. Microbiol. 2017, 55, 2996–3005. [Google Scholar] [CrossRef] [PubMed]
  13. Sherry, L.; Ramage, G.; Kean, R.; Borman, A.; Johnson, E.M.; Richardson, M.D.; Rautemaa-Richardson, R. Biofilm-Forming Capability of Highly Virulent, Multidrug-Resistant Candida auris. Emerg. Infect. Dis. 2017, 23, 328–331. [Google Scholar] [CrossRef] [PubMed]
  14. Ledwoch, K.; Maillard, J.Y. Candida auris Dry Surface Biofilm (DSB) for Disinfectant Efficacy Testing. Materials 2018, 12, 18. [Google Scholar] [CrossRef] [PubMed]
  15. Ruiz-Gaitán, A.; Martínez, H.; Moret, A.M.; Calabuig, E.; Tasias, M.; Alastruey-Izquierdo, A.; Zaragoza, Ó.; Mollar, J.; Frasquet, J.; Salavert-Lletí, M.; et al. Detection and Treatment of Candida auris in an Outbreak Situation: Risk Factors for Developing Colonization and Candidemia by This New Species in Critically Ill Patients. Expert Rev. Anti. Infect Ther. 2019, 17, 295–305. [Google Scholar] [CrossRef] [PubMed]
  16. Antifungal Susceptibility Testing and Interpretation|Candida auris|Fungal Diseases|CDC. Available online: https://www.cdc.gov/fungal/candida-auris/c-auris-antifungal.html (accessed on 8 May 2024).
  17. Lone, S.A.; Ahmad, A. Candida auris-the Growing Menace to Global Health. Mycoses 2019, 62, 620–637. [Google Scholar] [CrossRef] [PubMed]
  18. Ademe, M.; Girma, F. Candida auris: From Multidrug Resistance to Pan-Resistant Strains. Infect. Drug. Resist. 2020, 13, 1287–1294. [Google Scholar] [CrossRef] [PubMed]
  19. Chakrabarti, A.; Sood, P. On the Emergence, Spread and Resistance of Candida auris: Host, Pathogen and Environmental Tipping Points. J. Med. Microbiol. 2021, 70, 001318. [Google Scholar] [CrossRef] [PubMed]
  20. Lyman, M.; Forsberg, K.; Sexton, D.J.; Chow, N.A.; Lockhart, S.R.; Jackson, B.R.; Chiller, T. Worsening Spread of Candida auris in the United States, 2019 to 2021. Ann. Intern. Med. 2023, 176, 489–495. [Google Scholar] [CrossRef]
  21. Almaghrabi, R.S.; Albalawi, R.; Mutabagani, M.; Atienza, E.; Aljumaah, S.; Gade, L.; Forsberg, K.; Litvintseva, A.; Althawadi, S. Molecular Characterisation and Clinical Outcomes of Candida auris Infection: Single-Centre Experience in Saudi Arabia. Mycoses 2020, 63, 452–460. [Google Scholar] [CrossRef]
  22. Khan, Z.; Ahmad, S.; Al-Sweih, N.; Joseph, L.; Alfouzan, W.; Asadzadeh, M. Increasing Prevalence, Molecular Characterization and Antifungal Drug Susceptibility of Serial Candida auris Isolates in Kuwait. PLoS ONE 2018, 13, e0195743. [Google Scholar] [CrossRef] [PubMed]
  23. Al Maani, A.; Paul, H.; Al-Rashdi, A.; Al Wahaibi, A.; Al-Jardani, A.; Al Abri, A.M.A.; Al Balushi, M.A.H.; Al Abri, S.; Al Reesi, M.; Al Maqbali, A.; et al. Ongoing Challenges with Healthcare-Associated Candida auris Outbreaks in Oman. J. Fungi 2019, 5, 101. [Google Scholar] [CrossRef]
  24. Whaley, S.G.; Berkow, E.L.; Rybak, J.M.; Nishimoto, A.T.; Barker, K.S.; Rogers, P.D. Azole Antifungal Resistance in Candida albicans and Emerging Non- Albicans Candida Species. Front. Microbiol. 2017, 7, 231851. [Google Scholar] [CrossRef]
  25. Rybak, J.M.; Muñoz, J.F.; Barker, K.S.; Parker, J.E.; Esquivel, B.D.; Berkow, E.L.; Lockhart, S.R.; Gade, L.; Palmer, G.E.; White, T.C.; et al. Mutations in TAC1B: A Novel Genetic Determinant of Clinical Fluconazole Resistance in Candida auris. mBio 2020, 11, e00365-20. [Google Scholar] [CrossRef]
  26. Rybak, J.M.; Barker, K.S.; Muñoz, J.F.; Parker, J.E.; Ahmad, S.; Mokaddas, E.; Abdullah, A.; Elhagracy, R.S.; Kelly, S.L.; Cuomo, C.A.; et al. In Vivo Emergence of High-Level Resistance during Treatment Reveals the First Identified Mechanism of Amphotericin B Resistance in Candida auris. Clin. Microbiol. Infect. 2022, 28, 838–843. [Google Scholar] [CrossRef]
  27. Biagi, M.J.; Wiederhold, N.P.; Gibas, C.; Wickes, B.L.; Lozano, V.; Bleasdale, S.C.; Danziger, L. Development of High-Level Echinocandin Resistance in a Patient with Recurrent Candida auris Candidemia Secondary to Chronic Candiduria. In Open Forum Infectious Diseases; Oxford University Press: Oxford, UK, 2019; Volume 6. [Google Scholar] [CrossRef]
  28. Kordalewska, M.; Lee, A.; Park, S.; Berrio, I.; Chowdhary, A.; Zhao, Y.; Perlin, D.S. Understanding Echinocandin Resistance in the Emerging Pathogen Candida auris. Antimicrob. Agents Chemother. 2018, 62, e00238-18. [Google Scholar] [CrossRef]
  29. Rybak, J.M.; Cuomo, C.A.; David Rogers, P. The Molecular and Genetic Basis of Antifungal Resistance in the Emerging Fungal Pathogen Candida auris. Curr. Opin. Microbiol. 2022, 70, 102208. [Google Scholar] [CrossRef]
  30. Berkow, E.L.; Lockhart, S.R. Activity of CD101, a Long-Acting Echinocandin, against Clinical Isolates of Candida auris. Diagn. Microbiol. Infect Dis. 2018, 90, 196–197. [Google Scholar] [CrossRef]
  31. Wang, S.; Pan, J.; Gu, L.; Wang, W.; Wei, B.; Zhang, H.; Chen, J.; Wang, H. Review of Treatment Options for a Multidrug-Resistant Fungus: Candida auris. Med. Mycol. 2024, 62, myad127. [Google Scholar] [CrossRef]
  32. Pfaller, M.A.; Messer, S.A.; Rhomberg, P.R.; Borroto-Esoda, K.; Castanheira, M. Differential Activity of the Oral Glucan Synthase Inhibitor SCY-078 against Wild-Type and Echinocandin-Resistant Strains of Candida Species. Antimicrob. Agents Chemother. 2017, 61, e00161-17. [Google Scholar] [CrossRef]
  33. Wiederhold, N.P.; Najvar, L.K.; Olivo, M.; Morris, K.N.; Patterson, H.P.; Catano, G.; Patterson, T.F. Ibrexafungerp Demonstrates In Vitro Activity against Fluconazole-Resistant Candida auris and In Vivo Efficacy with Delayed Initiation of Therapy in an Experimental Model of Invasive Candidiasis. Antimicrob. Agents Chemother. 2021, 65, e02694-20. [Google Scholar] [CrossRef]
  34. Quindós, G.; Miranda-Cadena, K.; San-Millán, R.; Borroto-Esoda, K.; Cantón, E.; Linares-Sicilia, M.J.; Hamprecht, A.; Montesinos, I.; Tortorano, A.M.; Prigitano, A.; et al. In Vitro Antifungal Activity of Ibrexafungerp (SCY-078) Against Contemporary Blood Isolates from Medically Relevant Species of Candida: A European Study. Front. Cell Infect. Microbiol. 2022, 12, 906563. [Google Scholar] [CrossRef]
  35. Larkin, E.; Hager, C.; Chandra, J.; Mukherjee, P.K.; Retuerto, M.; Salem, I.; Long, L.; Isham, N.; Kovanda, L.; Borroto-Esoda, K.; et al. The Emerging Pathogen Candida auris: Growth Phenotype, Virulence Factors, Activity of Antifungals, and Effect of SCY-078, a Novel Glucan Synthesis Inhibitor, on Growth Morphology and Biofilm Formation. Antimicrob. Agents Chemother. 2017, 61, e02396-16. [Google Scholar] [CrossRef]
  36. Berkow, E.L.; Angulo, D.; Lockhart, S.R. In Vitro Activity of a Novel Glucan Synthase Inhibitor, SCY-078, against Clinical Isolates of Candida auris. Antimicrob. Agents Chemother. 2017, 61, e00435-17. [Google Scholar] [CrossRef]
  37. Hodges, M.R.; Ople, E.; Wedel, P.; Shaw, K.J.; Jakate, A.; Kramer, W.G.; van Marle, S.; van Hoogdalem, E.J.; Tawadrous, M. Safety and Pharmacokinetics of Intravenous and Oral Fosmanogepix, a First-in-Class Antifungal Agent, in Healthy Volunteers. Antimicrob. Agents Chemother. 2023, 67, e01623-22. [Google Scholar] [CrossRef]
  38. Pappas, P.G.; Vazquez, J.A.; Oren, I.; Rahav, G.; Aoun, M.; Bulpa, P.; Ben-Ami, R.; Ferrer, R.; Mccarty, T.; Thompson, G.R.; et al. Clinical Safety and Efficacy of Novel Antifungal, Fosmanogepix, for the Treatment of Candidaemia: Results from a Phase 2 Trial. J. Antimicrob. Chemother. 2023, 78, 2471–2480. [Google Scholar] [CrossRef]
  39. Vahedi-Shahandashti, R.; Lass-Flörl, C. Novel Antifungal Agents and Their Activity against Aspergillus Species. J. Fungi 2020, 6, 213. [Google Scholar] [CrossRef]
  40. Wiederhold, N.P. Review of T-2307, an Investigational Agent That Causes Collapse of Fungal Mitochondrial Membrane Potential. J. Fungi 2021, 7, 130. [Google Scholar] [CrossRef]
  41. Santana, D.J.; Anku, J.A.E.; Zhao, G.; Zarnowski, R.; Johnson, C.J.; Hautau, H.; Visser, N.D.; Ibrahim, A.S.; Andes, D.; Nett, J.E.; et al. A Candida auris-Specific Adhesin, Scf1, Governs Surface Association, Colonization, and Virulence. Science 2023, 381, 1461–1467. [Google Scholar] [CrossRef]
  42. Toepfer, S.; Lackner, M.; Keniya, M.V.; Zenz, L.M.; Friemert, M.; Bracher, F.; Monk, B.C. Clorgyline Analogs Synergize with Azoles against Drug Efflux in Candida auris. J. Fungi 2023, 9, 663. [Google Scholar] [CrossRef]
  43. Chi, X.; Zhang, H.; Wu, H.; Li, X.; Li, L.; Jiang, Y.; Ni, T. Discovery of Novel Tetrazoles Featuring a Pyrazole Moiety as Potent and Highly Selective Antifungal Agents. ACS Omega 2023, 8, 17103–17115. [Google Scholar] [CrossRef]
  44. Ni, T.; Chi, X.; Xie, F.; Li, L.; Wu, H.; Hao, Y.; Wang, X.; Zhang, D.; Jiang, Y. Design, Synthesis, and Evaluation of Novel Tetrazoles Featuring Isoxazole Moiety as Highly Selective Antifungal Agents. Eur. J. Med. Chem. 2023, 246, 115007. [Google Scholar] [CrossRef]
  45. Tu, J.; Zhu, T.; Wang, Q.; Yang, W.; Huang, Y.; Xu, D.; Liu, N.; Sheng, C. Discovery of a New Chemical Scaffold for the Treatment of Superbug Candida auris Infections. Emerg. Microbes Infect. 2023, 12, 2208687. [Google Scholar] [CrossRef]
  46. Li, J.; Coste, A.T.; Bachmann, D.; Sanglard, D.; Lamoth, F. Assessment of the In Vitro and In Vivo Antifungal Activity of NSC319726 against Candida auris. Microbiol. Spectr. 2021, 9, e01395-21. [Google Scholar] [CrossRef] [PubMed]
  47. Srivastava, V.; Wani, M.Y.; Al-Bogami, A.S.; Ahmad, A. Piperidine Based 1,2,3-Triazolylacetamide Derivatives Induce Cell Cycle Arrest and Apoptotic Cell Death in Candida auris. J. Adv. Res. 2020, 29, 121–135. [Google Scholar] [CrossRef]
  48. Wani, M.Y.; Alghamidi, M.S.S.; Srivastava, V.; Ahmad, A.; Aqlan, F.M.; Al-Bogami, A.S. Click Synthesis of Pyrrolidine-Based 1,2,3-Triazole Derivatives as Antifungal Agents Causing Cell Cycle Arrest and Apoptosis in Candida auris. Bioorg. Chem. 2023, 136, 106562. [Google Scholar] [CrossRef] [PubMed]
  49. Fuchs, F.; Hof, H.; Hofmann, S.; Kurzai, O.; Meis, J.F.; Hamprecht, A. Antifungal Activity of Nitroxoline against Candida auris Isolates. Clin. Microbiol. Infect. 2021, 27, 1697.e7–1697.e10. [Google Scholar] [CrossRef] [PubMed]
  50. Maphanga, T.G.; Mpembe, R.S.; Naicker, S.D.; Govender, N.P. In Vitro Antifungal Activity of Manogepix and Other Antifungal Agents against South African Candida auris Isolates from Bloodstream Infections. Microbiol. Spectr. 2022, 10, e01717-21. [Google Scholar] [CrossRef]
  51. Zhu, Y.C.; Kilburn, S.; Kapoor, M.; Chaturvedi, S.; Shaw, K.J.; Chaturvedi, V. In Vitro Activity of Manogepix against Multidrug-Resistant and Panresistant Candida auris from the New York Outbreak. Antimicrob. Agents Chemother. 2020, 64, e01124-20. [Google Scholar] [CrossRef]
  52. Dennis, E.K.; Kim, J.H.; Parkin, S.; Awuah, S.G.; Garneau-Tsodikova, S. Distorted Gold(I)-Phosphine Complexes as Antifungal Agents. J. Med. Chem. 2020, 63, 2455–2469. [Google Scholar] [CrossRef]
  53. Mohammad, H.; Eldesouky, H.E.; Hazbun, T.; Mayhoub, A.S.; Seleem, M.N. Identification of a Phenylthiazole Small Molecule with Dual Antifungal and Antibiofilm Activity Against Candida albicans and Candida auris. Sci. Rep. 2019, 9, 18941. [Google Scholar] [CrossRef] [PubMed]
  54. Hashemi, M.M.; Rovig, J.; Holden, B.S.; Taylor, M.F.; Weber, S.; Wilson, J.; Hilton, B.; Zaugg, A.L.; Ellis, S.W.; Yost, C.D.; et al. Ceragenins Are Active against Drug-Resistant Candida auris Clinical Isolates in Planktonic and Biofilm Forms. J. Antimicrob. Chemother. 2018, 73, 1537–1545. [Google Scholar] [CrossRef] [PubMed]
  55. John, L.L.H.; Thomson, D.D.; Bicanic, T.; Hoenigl, M.; Brown, A.J.P.; Harrison, T.S.; Bignell, E.M. Heightened Efficacy of Anidulafungin When Used in Combination with Manogepix or 5-Flucytosine against Candida auris In Vitro. Antimicrob. Agents Chemother. 2023, 67, e01645-22. [Google Scholar] [CrossRef] [PubMed]
  56. Parker, R.A.; Gabriel, K.T.; Graham, K.D.; Butts, B.K.; Cornelison, C.T. Antifungal Activity of Select Essential Oils against Candida auris and Their Interactions with Antifungal Drugs. Pathogens 2022, 11, 821. [Google Scholar] [CrossRef] [PubMed]
  57. Balkrishna, A.; Kharayat, B.; Rastogi, S.; Kabdwal, M.; Haldar, S.; Varshney, A. Withania Somnifera Seed Oil Exhibits Antibiofilm Properties against Drug-Resistant Candida auris Clinical Isolate through Modulation in Cell Permeability. J. Appl. Microbiol. 2023, 134, lxad087. [Google Scholar] [CrossRef] [PubMed]
  58. Di Vito, M.; Garzoli, S.; Rosato, R.; Mariotti, M.; Gervasoni, J.; Santucci, L.; Ovidi, E.; Cacaci, M.; Lombarini, G.; Torelli, R.; et al. A New Potential Resource in the Fight against Candida auris: The Cinnamomum Zeylanicum Essential Oil in Synergy with Antifungal Drug. Microbiol. Spectr. 2023, 11, e04385-22. [Google Scholar] [CrossRef]
  59. Rosato, R.; Napoli, E.; Granata, G.; Di Vito, M.; Garzoli, S.; Geraci, C.; Rizzo, S.; Torelli, R.; Sanguinetti, M.; Bugli, F. Study of the Chemical Profile and Anti-Fungal Activity against Candida auris of Cinnamomum Cassia Essential Oil and of Its Nano-Formulations Based on Polycaprolactone. Plants 2023, 12, 358. [Google Scholar] [CrossRef] [PubMed]
  60. Shreaz, S.; Wani, W.A.; Behbehani, J.M.; Raja, V.; Irshad, M.; Karched, M.; Ali, I.; Siddiqi, W.A.; Hun, L.T. Cinnamaldehyde and Its Derivatives, a Novel Class of Antifungal Agents. Fitoterapia 2016, 112, 116–131. [Google Scholar] [CrossRef] [PubMed]
  61. Doyle, A.A.; Stephens, J.C. A Review of Cinnamaldehyde and Its Derivatives as Antibacterial Agents. Fitoterapia 2019, 139, 104405. [Google Scholar] [CrossRef]
  62. Zhang, F.; Zhao, M.; Braun, D.R.; Ericksen, S.S.; Piotrowski, J.S.; Nelson, J.; Peng, J.; Ananiev, G.E.; Chanana, S.; Barns, K.; et al. A Marine Microbiome Antifungal Targets Urgent-Threat Drug-Resistant Fungi. Science 2020, 370, 974–978. [Google Scholar] [CrossRef]
  63. Marquez, L.; Lee, Y.; Duncan, D.; Whitesell, L.; Cowen, L.E.; Quave, C. Potent Antifungal Activity of Penta-O-Galloyl-β-d-Glucose against Drug-Resistant Candida albicans, Candida auris, and Other Non-albicans Candida Species. ACS Infect Dis. 2023, 9, 1685–1694. [Google Scholar] [CrossRef] [PubMed]
  64. Cheng, Z.; Wu, W.; Liu, Y.; Chen, S.; Li, H.; Yang, X.; Zhu, X.; Chen, X.; Yan, L.; Chu, Z.; et al. Natural Enfumafungin Analogues from Hormonema carpetanum and Their Antifungal Activities. J. Nat. Prod. 2023, 86, 2407–2413. [Google Scholar] [CrossRef] [PubMed]
  65. Du, L.; Haldar, S.; King, J.B.; Mattes, A.O.; Srivastava, S.; Wendt, K.L.; You, J.; Cunningham, C.; Cichewicz, R.H. Persephacin Is a Broad-Spectrum Antifungal Aureobasidin Metabolite That Overcomes Intrinsic Resistance in Aspergillus Fumigatus. J. Nat. Prod. 2023, 86, 1980–1993. [Google Scholar] [CrossRef] [PubMed]
  66. Jagels, A.; Adpressa, D.A.; Kaweesa, E.N.; McCauley, M.; Philmus, B.; Strother, J.A.; Loesgen, S. Metabolomics-Guided Discovery, Isolation, Structure Elucidation, and Bioactivity of Myropeptins C-E from Myrothecium inundatum. J. Nat. Prod. 2023, 86, 1723–1735. [Google Scholar] [CrossRef] [PubMed]
  67. Watanabe, Y.; Yoshida, Y.; Tokiwa, T.; Higo, M.; Ban, S.; Ikeda, A.; Noguchi, Y.; Hirose, T.; Sunazuka, T.; Nonaka, K.; et al. Hakuhybotric Acid, a New Antifungal Polyketide Produced by a Mycoparasitic Fungus Hypomyces pseudocorticiicola FKI-9008. J. Gen. Appl. Microbiol. 2022, 68, 200–206. [Google Scholar] [CrossRef] [PubMed]
  68. Rossoni, R.D.; de Barros, P.P.; do Carmo Mendonça, I.; Medina, R.P.; Silva, D.H.S.; Fuchs, B.B.; Junqueira, J.C.; Mylonakis, E. The Postbiotic Activity of Lactobacillus paracasei 28.4 Against Candida auris. Front. Cell Infect. Microbiol. 2020, 10, 397. [Google Scholar] [CrossRef] [PubMed]
  69. Ismail, M.; Srivastava, V.; Marimani, M.; Ahmad, A. Carvacrol Modulates the Expression and Activity of Antioxidant Enzymes in Candida auris. Res. Microbiol. 2022, 173, 103916. [Google Scholar] [CrossRef] [PubMed]
  70. Fatima, T.; Fatima, Z.; Hameed, S. Abrogation of Efflux Pump Activity, Biofilm Formation, and Immune Escape by Candidacidal Geraniol in Emerging Superbug, Candida auris. Int. Microbiol. 2023, 26, 881–891. [Google Scholar] [CrossRef]
  71. Bentz, M.L.; Nunnally, N.; Lockhart, S.R.; Sexton, D.J.; Berkow, E.L. Antifungal Activity of Nikkomycin Z against Candida auris. J. Antimicrob. Chemother. 2021, 76, 1495–1497. [Google Scholar] [CrossRef]
  72. Kim, H.R.; Eom, Y.B. Antifungal and Anti-Biofilm Effects of 6-Shogaol against Candida auris. J. Appl. Microbiol. 2021, 130, 1142–1153. [Google Scholar] [CrossRef]
  73. Sharifi-Rad, M.; Varoni, E.M.; Iriti, M.; Martorell, M.; Setzer, W.N.; del Mar Contreras, M.; Salehi, B.; Soltani-Nejad, A.; Rajabi, S.; Tajbakhsh, M.; et al. Carvacrol and Human Health: A Comprehensive Review. Phytother. Res. 2018, 32, 1675–1687. [Google Scholar] [CrossRef] [PubMed]
  74. Ok, S.; Jeong, W.S. Optimization of Extraction Conditions for the 6-Shogaol-Rich Extract from Ginger (Zingiber Officinale Roscoe). Prev. Nutr. Food Sci. 2012, 17, 166–171. [Google Scholar] [CrossRef] [PubMed]
  75. Zeng, H.; Stadler, M.; Abraham, W.R.; Müsken, M.; Schrey, H. Inhibitory Effects of the Fungal Pigment Rubiginosin C on Hyphal and Biofilm Formation in Candida albicans and Candida auris. J. Fungi 2023, 9, 726. [Google Scholar] [CrossRef] [PubMed]
  76. Zhao, M.; Zhang, F.; Zarnowski, R.; Barns, K.; Jones, R.; Fossen, J.; Sanchez, H.; Rajski, S.R.; Audhya, A.; Bugni, T.S.; et al. Turbinmicin Inhibits Candida Biofilm Growth by Disrupting Fungal Vesicle-Mediated Trafficking. J. Clin. Investig. 2021, 131, 1–5. [Google Scholar] [CrossRef] [PubMed]
  77. Antimicrobial Peptide Database. Available online: https://aps.unmc.edu/ (accessed on 8 May 2024).
  78. Perez-Rodriguez, A.; Eraso, E.; Quindós, G.; Mateo, E. Antimicrobial Peptides with Anti-Candida Activity. Int. J. Mol. Sci. 2022, 23, 9264. [Google Scholar] [CrossRef] [PubMed]
  79. Tóth, L.; Kele, Z.; Borics, A.; Nagy, L.G.; Váradi, G.; Virágh, M.; Takó, M.; Vágvölgyi, C.; Galgóczy, L. NFAP2, a Novel Cysteine-Rich Anti-Yeast Protein from Neosartorya fischeri NRRL 181: Isolation and Characterization. AMB Express. 2016, 6, 75. [Google Scholar] [CrossRef] [PubMed]
  80. Kovács, L.; Virágh, M.; Takó, M.; Papp, T.; Vágvölgyi, C.; Galgóczy, L. Isolation and Characterization of Neosartorya fischeri Antifungal Protein (NFAP). Peptides 2011, 32, 1724–1731. [Google Scholar] [CrossRef] [PubMed]
  81. Kovács, R.; Holzknecht, J.; Hargitai, Z.; Papp, C.; Farkas, A.; Borics, A.; Tóth, L.; Váradi, G.; Tóth, G.K.; Kovács, I.; et al. In Vivo Applicability Neosartorya fischeri Antifungal Protein 2 (NFAP2) in Treatment of Vulvovaginal Candidiasis. Antimicrob. Agents Chemother. 2019, 63, e01777-18. [Google Scholar] [CrossRef] [PubMed]
  82. Kovács, R.; Nagy, F.; Tóth, Z.; Forgács, L.; Tóth, L.; Váradi, G.; Tóth, G.K.; Vadászi, K.; Borman, A.M.; Majoros, L.; et al. The Neosartorya fischeri Antifungal Protein 2 (NFAP2): A New Potential Weapon against Multidrug-Resistant Candida auris Biofilms. Int. J. Mol. Sci. 2021, 22, 771. [Google Scholar] [CrossRef]
  83. Ramesh, S.; Madduri, M.; Rudramurthy, S.M.; Roy, U. Functional Characterization of a Bacillus-Derived Novel Broad-Spectrum Antifungal Lipopeptide Variant against Candida tropicalis and Candida auris and Unravelling Its Mode of Action. Microbiol. Spectr. 2023, 11, e01583-22. [Google Scholar] [CrossRef]
  84. Shaban, S.; Patel, M.; Ahmad, A. Fungicidal Activity of Human Antimicrobial Peptides and Their Synergistic Interaction with Common Antifungals against Multidrug-Resistant Candida auris. Int. Microbiol. 2023, 26, 165–177. [Google Scholar] [CrossRef]
  85. Rather, I.A.; Sabir, J.S.M.; Asseri, A.H.; Ali, S. Antifungal Activity of Human Cathelicidin LL-37, a Membrane Disrupting Peptide, by Triggering Oxidative Stress and Cell Cycle Arrest in Candida auris. J. Fungi 2022, 8, 204. [Google Scholar] [CrossRef]
  86. Pathirana, R.U.; Friedman, J.; Norris, H.L.; Salvatori, O.; McCall, A.D.; Kay, J.; Edgerton, M. Fluconazole-Resistant Candida auris Is Susceptible to Salivary Histatin 5 Killing and to Intrinsic Host Defenses. Antimicrob. Agents Chemother. 2018, 62, e01872-17. [Google Scholar] [CrossRef]
  87. Kamli, M.R.; Sabir, J.S.M.; Malik, M.A.; Ahmad, A. Characterization of Defensin-like Protein 1 for Its Anti-Biofilm and Anti-Virulence Properties for the Development of Novel Antifungal Drug against Candida auris. J. Fungi. 2022, 8, 1298. [Google Scholar] [CrossRef]
  88. Dal Mas, C.; Rossato, L.; Shimizu, T.; Oliveira, E.B.; Da Silva Junior, P.I.; Meis, J.F.; Hayashi, M.A.F.; Colombo, A.L. Effects of the Natural Peptide Crotamine from a South American Rattlesnake on Candida auris, an Emergent Multidrug Antifungal Resistant Human Pathogen. Biomolecules 2019, 9, 205. [Google Scholar] [CrossRef]
  89. Raber, H.F.; Sejfijaj, J.; Kissmann, A.K.; Wittgens, A.; Gonzalez-Garcia, M.; Alba, A.; Vázquez, A.A.; Vicente, F.E.M.; Erviti, J.P.; Kubiczek, D.; et al. Antimicrobial Peptides Pom-1 and Pom-2 from Pomacea poeyana Are Active against Candida auris, C. Parapsilosis and C. Albicans Biofilms. Pathogens 2021, 10, 496. [Google Scholar] [CrossRef]
  90. Pinheiro, T.K.S.; Nicola, A.M.; Bentes, D.B.; Zhang, S.X.; Felipe, M.S.S.; Silva-Pereira, I.; Albuquerque, P. The Antimicrobial Peptide ToAP2 Is Synergic with Caspofungin and Amphotericin B against Candida auris. preprint 2023. [Google Scholar] [CrossRef]
  91. dos Reis, T.F.; de Castro, P.A.; Bastos, R.W.; Pinzan, C.F.; Souza, P.F.N.; Ackloo, S.; Hossain, M.A.; Drewry, D.H.; Alkhazraji, S.; Ibrahim, A.S.; et al. A Host Defense Peptide Mimetic, Brilacidin, Potentiates Caspofungin Antifungal Activity against Human Pathogenic Fungi. Nat. Commun. 2023, 14, 2052. [Google Scholar] [CrossRef]
  92. Bugli, F.; Massaro, F.; Buonocore, F.; Saraceni, P.R.; Borocci, S.; Ceccacci, F.; Bombelli, C.; Di Vito, M.; Marchitiello, R.; Mariotti, M.; et al. Design and Characterization of Myristoylated and Non-Myristoylated Peptides Effective against Candida Spp. Clinical Isolates. Int. J. Mol. Sci. 2022, 23, 2164. [Google Scholar] [CrossRef]
  93. Szerencsés, B.; Gácser, A.; Endre, G.; Domonkos, I.; Tiricz, H.; Vágvölgyi, C.; Szolomajer, J.; Howan, D.H.O.; Tóth, G.K.; Pfeiffer, I.; et al. Symbiotic NCR Peptide Fragments Affect the Viability, Morphology and Biofilm Formation of Candida Species. Int. J. Mol. Sci. 2021, 22, 3666. [Google Scholar] [CrossRef]
  94. Vicente, F.E.M.; González-Garcia, M.; Diaz Pico, E.; Moreno-Castillo, E.; Garay, H.E.; Rosi, P.E.; Jimenez, A.M.; Campos-Delgado, J.A.; Rivera, D.G.; Chinea, G.; et al. Design of a Helical-Stabilized, Cyclic, and Nontoxic Analogue of the Peptide Cm-P5 with Improved Antifungal Activity. ACS Omega 2019, 4, 19081–19095. [Google Scholar] [CrossRef]
  95. Basso, V.; Garcia, A.; Tran, D.Q.; Schaal, J.B.; Tran, P.; Ngole, D.; Aqeel, Y.; Tongaonkar, P.; Ouellette, A.J.; Selsteda, M.E. Fungicidal Potency and Mechanisms of θ-Defensins against Multidrug-Resistant Candida Species. Antimicrob. Agents Chemother. 2018, 62, e00111-18. [Google Scholar] [CrossRef] [PubMed]
  96. Nikawa, H.; Jin, C.; Fukushima, H.; Makihira, S.; Hamada, T. Antifungal Activity of Histatin-5 against Non-albicans Candida Species. Oral Microbiol. Immunol. 2001, 16, 250–252. [Google Scholar] [CrossRef]
  97. Peters, B.M.; Zhu, J.; Fidel, P.L.; Scheper, M.A.; Hackett, W.; El Shaye, S.; Jabra-Rizk, M.A. Protection of the Oral Mucosa by Salivary Histatin-5 against Candida albicans in an Ex Vivo Murine Model of Oral Infection. FEMS Yeast Res. 2010, 10, 597–604. [Google Scholar] [CrossRef]
  98. Liao, H.; Liu, S.; Wang, H.; Su, H.; Liu, Z. Efficacy of Histatin5 in a Murine Model of Vulvovaginal Candidiasis Caused by Candida albicans. Pathog. Dis. 2017, 75, ftx072. [Google Scholar] [CrossRef]
  99. Aerts, A.M.; Bammens, L.; Govaert, G.; Carmona-Gutierrez, D.; Madeo, F.; Cammue, B.P.A.; Thevissen, K. The Antifungal Plant Defensin HsAFP1 from Heuchera sanguinea Induces Apoptosis in Candida albicans. Front. Microbiol. 2011, 2, 47. [Google Scholar] [CrossRef]
  100. Hayes, B.M.E.; Bleackley, M.R.; Wiltshire, J.L.; Anderson, M.A.; Traven, A.; Van Der Weerden, N.L. Identification and Mechanism of Action of the Plant Defensin NaD1 as a New Member of the Antifungal Drug Arsenal against Candida albicans. Antimicrob. Agents Chemother. 2013, 57, 3667–3675. [Google Scholar] [CrossRef] [PubMed]
  101. Gonçalves, S.; Silva, P.M.; Felício, M.R.; de Medeiros, L.N.; Kurtenbach, E.; Santos, N.C. Ps D1 Effects on Candida albicans Planktonic Cells and Biofilms. Front. Cell Infect. Microbiol. 2017, 7, 249. [Google Scholar] [CrossRef] [PubMed]
  102. García, M.G.; Rodríguez, A.; Alba, A.; Vázquez, A.A.; Vicente, F.E.M.; Pérez-Erviti, J.; Spellerberg, B.; Stenger, S.; Grieshober, M.; Conzelmann, C.; et al. New Antibacterial Peptides from the Freshwater Mollusk Pomacea poeyana (Pilsbry, 1927). Biomolecules 2020, 10, 1473. [Google Scholar] [CrossRef]
  103. Dekkerová, J.; Lopez-Ribot, J.L.; Bujdáková, H. Activity of Anti-CR3-RP Polyclonal Antibody against Biofilms Formed by Candida auris, a Multidrug-Resistant Emerging Fungal Pathogen. Eur. J. Clin. Microbiol. Infect Dis. 2019, 38, 101–108. [Google Scholar] [CrossRef]
  104. Singh, S.; Barbarino, A.; Youssef, E.G.; Coleman, D.; Gebremariam, T.; Ibrahim, A.S. Protective Efficacy of Anti-Hyr1p Monoclonal Antibody against Systemic Candidiasis Due to Multi-Drug-Resistant Candida auris. J. Fungi 2023, 9, 103. [Google Scholar] [CrossRef] [PubMed]
  105. Singh, S.; Uppuluri, P.; Mamouei, Z.; Alqarihi, A.; Elhassan, H.; French, S.; Lockhart, S.R.; Chiller, T.; Edwards, J.E.; Ibrahim, A.S. The NDV-3A Vaccine Protects Mice from Multidrug Resistant Candida auris Infection. PLoS Pathog. 2019, 15, e1007460. [Google Scholar] [CrossRef] [PubMed]
  106. Di Mambro, T.; Vanzolini, T.; Bruscolini, P.; Perez-Gaviro, S.; Marra, E.; Roscilli, G.; Bianchi, M.; Fraternale, A.; Schiavano, G.F.; Canonico, B.; et al. A New Humanized Antibody Is Effective against Pathogenic Fungi in Vitro. Sci. Rep. 2021, 11, 19500. [Google Scholar] [CrossRef] [PubMed]
  107. Vanzolini, T.; Di Mambro, T.; Magnani, M.; Menotta, M. AFM Evaluation of a Humanized Recombinant Antibody Affecting C. Auris Cell Wall and Stability. RSC Adv. 2023, 13, 6130–6142. [Google Scholar] [CrossRef] [PubMed]
  108. Rosario-colon, J.; Eberle, K.; Adams, A.; Courville, E.; Xin, H. Candida Cell-Surface-Specific Monoclonal Antibodies Protect Mice against Candida auris Invasive Infection. Int. J. Mol. Sci. 2021, 22, 6162. [Google Scholar] [CrossRef] [PubMed]
  109. Ben-Nathan, D.; Lustig, S.; Tam, G.; Robinzon, S.; Segal, S.; Rager-Zisman, B. Prophylactic and Therapeutic Efficacy of Human Intravenous Immunoglobulin in Treating West Nile Virus Infection in Mice. J. Infect. Dis. 2003, 188, 5–12. [Google Scholar] [CrossRef] [PubMed]
  110. Bayry, J.; Lacroix-Desmazes, S.; Kazatchkine, M.D.; Kaveri, S.V. Intravenous Immunoglobulin for Infectious Diseases: Back to the Pre-Antibiotic and Passive Prophylaxis Era? Trends Pharmacol. Sci. 2004, 25, 306–310. [Google Scholar] [CrossRef]
  111. Diep, B.A.; Le, V.T.M.; Badiou, C.; Le, H.N.; Pinheiro, M.G.; Duong, A.H.; Wang, X.; Dip, E.C.; Aguiar-Alves, F.; Basuino, L.; et al. IVIG-Mediated Protection against Necrotizing Pneumonia Caused by MRSA. Sci. Transl. Med. 2016, 8, 357ra124. [Google Scholar] [CrossRef] [PubMed]
  112. Xin, H.; Rosario-Colon, J.A.; Eberle, K. Novel Intravenous Immunoglobulin Therapy for the Prevention and Treatment of Candida auris and Candida albicans Disseminated Candidiasis. mSphere 2023, 8, e00584-22. [Google Scholar] [CrossRef]
  113. Khan, T.; Suleman, M.; Ali, S.S.; Sarwar, M.F.; Ali, I.; Ali, L.; Khan, A.; Rokhan, B.; Wang, Y.; Zhao, R.; et al. Subtractive Proteomics Assisted Therapeutic Targets Mining and Designing Ensemble Vaccine against Candida auris for Immune Response Induction. Comput. Biol. Med. 2022, 145, 105462. [Google Scholar] [CrossRef]
  114. Gupta, S.K.; Osmanoglu, Ö.; Minocha, R.; Bandi, S.R.; Bencurova, E.; Srivastava, M.; Dandekar, T. Genome-Wide Scan for Potential CD4+ T-Cell Vaccine Candidates in Candida auris by Exploiting Reverse Vaccinology and Evolutionary Information. Front. Med. 2022, 9, 1008527. [Google Scholar] [CrossRef] [PubMed]
  115. Grizante Barião, P.H.; Tonani, L.; Brancini, G.T.P.; Nascimento, E.; Braga, G.Ú.L.; Wainwright, M.; von Zeska Kress, M.R. In Vitro and in Vivo Photodynamic Efficacies of Novel and Conventional Phenothiazinium Photosensitizers against Multidrug-Resistant Candida auris. Photochem. Photobiol. Sci. 2022, 21, 1807–1818. [Google Scholar] [CrossRef] [PubMed]
  116. Rodrigues, G.B.; Dias-Baruffi, M.; Holman, N.; Wainwright, M.; Braga, G.U.L. In Vitro Photodynamic Inactivation of Candida Species and Mouse Fibroblasts with Phenothiazinium Photosensitisers and Red Light. Photodiagnosis Photodyn. Ther. 2013, 10, 141–149. [Google Scholar] [CrossRef]
  117. Bapat, P.S.; Nobile, C.J. Photodynamic Therapy Is Effective Against Candida auris Biofilms. Front. Cell Infect. Microbiol. 2021, 11, 713092. [Google Scholar] [CrossRef] [PubMed]
  118. Silva, A.R.; Cabral, F.V.; Silva, C.R.; Silva, D.F.T.; Freitas, A.Z.; Fontes, A.; Ribeiro, M.S. New Insights in Phenothiazinium-Mediated Photodynamic Inactivation of Candida auris. J. Fungi 2023, 9, 717. [Google Scholar] [CrossRef]
  119. Štefánek, M.; Černáková, L.; Dekkerová, J.; Bujdáková, H. Photodynamic Inactivation Effectively Eradicates Candida auris Biofilm despite Its Interference with the Upregulation of CDR1 and MDR1 Efflux Genes. J. Fungi. 2022, 8, 1137. [Google Scholar] [CrossRef]
  120. Capoci, I.R.G.; Faria, D.R.; Sakita, K.M.; Rodrigues-Vendramini, F.A.V.; Bonfim-Mendonça, P.D.S.; Becker, T.C.A.; Kioshima, É.S.; Svidzinski, T.I.E.; Maigret, B. Repurposing Approach Identifies New Treatment Options for Invasive Fungal Disease. Bioorg. Chem. 2019, 84, 87–97. [Google Scholar] [CrossRef]
  121. Kim, J.H.; Cheng, L.W.; Chan, K.L.; Tam, C.C.; Mahoney, N.; Friedman, M.; Shilman, M.M.; Land, K.M. Antifungal Drug Repurposing. Antibiotics 2020, 9, 812. [Google Scholar] [CrossRef]
  122. Pushpakom, S.; Iorio, F.; Eyers, P.A.; Escott, K.J.; Hopper, S.; Wells, A.; Doig, A.; Guilliams, T.; Latimer, J.; McNamee, C.; et al. Drug Repurposing: Progress, Challenges and Recommendations. Nat. Rev. Drug Discov. 2019, 18, 41–58. [Google Scholar] [CrossRef]
  123. Gowri, M.; Jayashree, B.; Jeyakanthan, J.; Girija, E.K. Sertraline as a Promising Antifungal Agent: Inhibition of Growth and Biofilm of Candida auris with Special Focus on the Mechanism of Action in Vitro. J. Appl. Microbiol. 2020, 128, 426–437. [Google Scholar] [CrossRef]
  124. Lohse, M.B.; Laurie, M.T.; Levan, S.; Ziv, N.; Ennis, C.L.; Nobile, C.J.; DeRisi, J.; Johnson, A.D. Broad Susceptibility of Candida auris Strains to 8-Hydroxyquinolines and Mechanisms of Resistance. mBio 2023, 14, e0137623. [Google Scholar] [CrossRef] [PubMed]
  125. Talevi, A.; Bellera, C.L. Challenges and Opportunities with Drug Repurposing: Finding Strategies to Find Alternative Uses of Therapeutics. Expert. Opin. Drug Discov. 2020, 15, 397–401. [Google Scholar] [CrossRef] [PubMed]
  126. Eldesouky, H.E.; Salama, E.A.; Li, X.; Hazbun, T.R.; Mayhoub, A.S.; Seleem, M.N. Repurposing Approach Identifies Pitavastatin as a Potent Azole Chemosensitizing Agent Effective against Azole-Resistant Candida Species. Sci. Rep. 2020, 10, 7525. [Google Scholar] [CrossRef] [PubMed]
  127. Eldesouky, H.E.; Lanman, N.A.; Hazbun, T.R.; Seleem, M.N. Aprepitant, an Antiemetic Agent, Interferes with Metal Ion Homeostasis of Candida auris and Displays Potent Synergistic Interactions with Azole Drugs. Virulence 2020, 11, 1466–1481. [Google Scholar] [CrossRef] [PubMed]
  128. Barreto, T.L.; Rossato, L.; de Freitas, A.L.D.; Meis, J.F.; Lopes, L.B.; Colombo, A.L.; Ishida, K. Miltefosine as an Alternative Strategy in the Treatment of the Emerging Fungus Candida auris. Int. J. Antimicrob. Agents 2020, 56, 106049. [Google Scholar] [CrossRef]
  129. Aghaei Gharehbolagh, S.; Izadi, A.; Talebi, M.; Sadeghi, F.; Zarrinnia, A.; Zarei, F.; Darmiani, K.; Borman, A.M.; Mahmoudi, S. New Weapons to Fight a New Enemy: A Systematic Review of Drug Combinations against the Drug-Resistant Fungus Candida auris. Mycoses 2021, 64, 1308–1316. [Google Scholar] [CrossRef] [PubMed]
  130. Bidaud, A.L.; Djenontin, E.; Botterel, F.; Chowdhary, A.; Dannaoui, E. Colistin Interacts Synergistically with Echinocandins against Candida auris. Int. J. Antimicrob. Agents 2020, 55, 105901. [Google Scholar] [CrossRef]
  131. Eldesouky, H.E.; Salama, E.A.; Lanman, N.A.; Hazbun, T.R.; Seleem, M.N. Potent Synergistic Interactions between Lopinavir and Azole Antifungal Drugs against Emerging Multidrug-Resistant Candida auris. Antimicrob. Agents. Chemother. 2020, 65, e00684-20. [Google Scholar] [CrossRef] [PubMed]
  132. Salama, E.A.; Eldesouky, H.E.; Elgammal, Y.; Abutaleb, N.S.; Seleem, M.N. Lopinavir and Ritonavir Act Synergistically with Azoles against Candida auris in Vitro and in a Mouse Model of Disseminated Candidiasis. Int. J. Antimicrob. Agents 2023, 62, 106906. [Google Scholar] [CrossRef]
  133. Elgammal, Y.; Salama, E.A.; Seleem, M.N. Atazanavir Resensitizes Candida auris to Azoles. Antimicrob. Agents. Chemother. 2023, 67, e01631-22. [Google Scholar] [CrossRef]
  134. Elgammal, Y.; Salama, E.A.; Seleem, M.N. Saquinavir Potentiates Itraconazole’s Antifungal Activity against Multidrug-Resistant Candida auris in Vitro Andin Vivo. Med. Mycol. 2023, 61, myad081. [Google Scholar] [CrossRef] [PubMed]
  135. Giacobbe, D.R.; Magnasco, L.; Sepulcri, C.; Mikulska, M.; Koehler, P.; Cornely, O.A.; Bassetti, M. Recent Advances and Future Perspectives in the Pharmacological Treatment of Candida auris Infections. Expert Rev. Clin. Pharmacol. 2021, 14, 1205–1220. [Google Scholar] [CrossRef] [PubMed]
  136. Hao, W.; Wang, Y.; Xi, Y.; Yang, Z.; Zhang, H.; Ge, X. Activity of Chlorhexidine Acetate in Combination with Fluconazole against Suspensions and Biofilms of Candida auris. J. Infect. Chemother. 2022, 28, 29–34. [Google Scholar] [CrossRef] [PubMed]
  137. Eldesouky, H.E.; Li, X.; Abutaleb, N.S.; Mohammad, H.; Seleem, M.N. Synergistic Interactions of Sulfamethoxazole and Azole Antifungal Drugs against Emerging Multidrug-Resistant Candida auris. Int. J. Antimicrob. Agents 2018, 52, 754–761. [Google Scholar] [CrossRef] [PubMed]
  138. Lara, H.H.; Ixtepan-Turrent, L.; Jose Yacaman, M.; Lopez-Ribot, J. Inhibition of Candida auris Biofilm Formation on Medical and Environmental Surfaces by Silver Nanoparticles. ACS Appl. Mater. Interfaces 2020, 12, 21183–21191. [Google Scholar] [CrossRef] [PubMed]
  139. Vazquez-Munoz, R.; Lopez, F.D.; Lopez-Ribot, J.L. Silver Nanoantibiotics Display Strong Antifungal Activity Against the Emergent Multidrug-Resistant Yeast Candida auris Under Both Planktonic and Biofilm Growing Conditions. Front. Microbiol. 2020, 11, 541606. [Google Scholar] [CrossRef] [PubMed]
  140. AlJindan, R.; AlEraky, D.M. Silver Nanoparticles: A Promising Antifungal Agent against the Growth and Biofilm Formation of the Emergent Candida auris. J. Fungi 2022, 8, 744. [Google Scholar] [CrossRef]
  141. Malik, M.A.; Batterjee, M.G.; Kamli, M.R.; Alzahrani, K.A.; Danish, E.Y.; Nabi, A. Polyphenol-Capped Biogenic Synthesis of Noble Metallic Silver Nanoparticles for Antifungal Activity against Candida auris. J. Fungi 2022, 8, 639. [Google Scholar] [CrossRef]
  142. Kamli, M.R.; Srivastava, V.; Hajrah, N.H.; Sabir, J.S.M.; Hakeem, K.R.; Ahmad, A.; Malik, M.A. Facile Bio-Fabrication of Ag-Cu-Co Trimetallic Nanoparticles and Its Fungicidal Activity against Candida auris. J. Fungi 2021, 7, 62. [Google Scholar] [CrossRef]
  143. Vazquez-Munoz, R.; Lopez, F.D.; Lopez-Ribot, J.L. Bismuth Nanoantibiotics Display Anticandidal Activity and Disrupt the Biofilm and Cell Morphology of the Emergent Pathogenic Yeast Candida auris. Antibiotics 2020, 9, 461. [Google Scholar] [CrossRef]
  144. Fayed, B.; Jayakumar, M.N.; Soliman, S.S.M. Caspofungin-Resistance in Candida auris Is Cell Wall-Dependent Phenotype and Potential Prevention by Zinc Oxide Nanoparticles. Med. Mycol. 2021, 59, 1243–1256. [Google Scholar] [CrossRef] [PubMed]
  145. de Alteriis, E.; Maione, A.; Falanga, A.; Bellavita, R.; Galdiero, S.; Albarano, L.; Salvatore, M.M.; Galdiero, E.; Guida, M. Activity of Free and Liposome-Encapsulated Essential Oil from Lavandula angustifolia against Persister-Derived Biofilm of Candida auris. Antibiotics 2021, 11, 26. [Google Scholar] [CrossRef] [PubMed]
  146. Baldim, I.; Paziani, M.H.; Grizante Barião, P.H.; von Zeska Kress, M.R.; Oliveira, W.P. Nanostructured Lipid Carriers Loaded with Lippia sidoides Essential Oil as a Strategy to Combat the Multidrug-Resistant Candida auris. Pharmaceutics 2022, 14, 180. [Google Scholar] [CrossRef] [PubMed]
  147. Jaromin, A.; Zarnowski, R.; Markowski, A.; Zagórska, A.; Johnson, C.J.; Etezadi, H.; Kihara, S.; Mota-Santiago, P.; Nett, J.E.; Boyd, B.J.; et al. Liposomal Formulation of a New Antifungal Hybrid Compound Provides Protection against Candida auris in the Ex Vivo Skin Colonization Model. Antimicrob. Agents Chemother. 2024, 68, e00955-23. [Google Scholar] [CrossRef] [PubMed]
Figure 1. An updated illustration of distribution pattern of C. auris in various geographical locations around the world. The data presented here are based on the information obtained from recent publications. A color scheme was used to represent the year of C. auris reports. Over 40 countries reported positive for C. auris between 2009 and 2022. By 2022, five clades had been reported worldwide; however, recently unpublished data reported the existence of a sixth clade, suspected to have originated in Singapore [9] (* represents the preprint of the publication).
Figure 1. An updated illustration of distribution pattern of C. auris in various geographical locations around the world. The data presented here are based on the information obtained from recent publications. A color scheme was used to represent the year of C. auris reports. Over 40 countries reported positive for C. auris between 2009 and 2022. By 2022, five clades had been reported worldwide; however, recently unpublished data reported the existence of a sixth clade, suspected to have originated in Singapore [9] (* represents the preprint of the publication).
Jof 10 00408 g001
Figure 2. Currently available treatment options for C. auris prophylaxis. As of December 2023, only one molecule was approved by US-FDA to manage C. auris. Rezafungin is a modified version of anidulafungin, with the structural modification aimed to reduce the hepatotoxicity of the molecule while its efficiency was retained. The data presented here are based on the recent information provided by Wang et al., 2024 [31].
Figure 2. Currently available treatment options for C. auris prophylaxis. As of December 2023, only one molecule was approved by US-FDA to manage C. auris. Rezafungin is a modified version of anidulafungin, with the structural modification aimed to reduce the hepatotoxicity of the molecule while its efficiency was retained. The data presented here are based on the recent information provided by Wang et al., 2024 [31].
Jof 10 00408 g002
Figure 3. Scheme of potential molecules from different sources of natural products against C. auris.
Figure 3. Scheme of potential molecules from different sources of natural products against C. auris.
Jof 10 00408 g003
Figure 4. List of commercial drug molecules used to manage other disease conditions showing antifungal action against C. auris.
Figure 4. List of commercial drug molecules used to manage other disease conditions showing antifungal action against C. auris.
Jof 10 00408 g004
Table 1. Minimal Inhibitory Concentration (MIC) of different classes of chemically derived compounds against C. auris.
Table 1. Minimal Inhibitory Concentration (MIC) of different classes of chemically derived compounds against C. auris.
GroupCompoundMIC or MIC RangeReference
Clorgyline analogsM1986.7–98.3 µMToepfer et al., 2023 [42]
M25137–228 µM
Novel tetrazoles featuring a pyrazole moiety8, 11, 15, 24, 25<0.0625–4, 0.125–8, <0.0625–2, 0.5–64, <0.0625–16 µg/mLChi et al., 2023 [43]
Novel tetrazoles featuring an isoxazole moiety10d, 10h, 13r, 13u0.008, <0.008, 0.0313, 0.0313 µg/mLNi et al., 2023 [44]
Benzoanilide groupCompound A10.5–2.0 µg/mLTu et al., 2023 [45]
Piperidine based 1,2,3-triazolylacetamide derivativespta1, pta2, pta3, pta4, pta5, pta6 0.48–0.97, 0.24–0.48, 0.12–0.24, >250, >250, >250 µg/mLSrivastava et al., 2020 [47]
Pyrrolidine-based 1,2,3-triazoleP1–P100.97–62.5 µg/mLWani et al., 2023 [48]
Nitroxoline 0.125 to 1 µg/mLFuchs et al., 2021 [49]
NSC319726, a thiosemicarbazone 0.125 to 0.25 µg/mLLi et al., 2021 [46]
ManogepixFluR0.002 to 0.063 µg/mLMaphanga et al., 2022 [50]
Flu and AmBR0.004 to 0.031 µg/mL
PanR0.004 and 0.008 µg/mL
ManogepixPanR—New York0.008 to 0.015 µg/mLZhu et al., 2020 [51]
Gold(I)—Phosphine Complex 4 3.9 to 7.8 µg/mLDennis et al., 2019 [52]
Gold(I)—Phosphine Complex 6 1.95 µg/mL
Phenylthiazole—Compound 1 0.25–2 µg/mLMohammad et al., 2019 [53]
CerageninsCSA-44, CSA-131, CSA-142, CSA-1440.5–1, 0.5–1, 2 to 8, 0.5–2 µg/mLHashemi et al., 2018 [54]
FluR—Fluconazole-resistant; AmBR—Amphotericin B-resistant; PanR—Resistant to more than two antifungals.
Table 2. Antifungal natural products against C. auris.
Table 2. Antifungal natural products against C. auris.
SourceMoleculeMIC/MIC RangeReference
Turbinmicin-producing bacterium Micromonospora sp. WMMC-415Turbinmicin0.125–0.50 µg/mLZhang et al., 2020 [62]
Hypoxylon rubiginosum and Hypoxylon texensePenta-O-galloyl-β-D-Glucose1–8 µg/mLMarquez et al., 2023 [63]
Hormonema carpetanumEnfumafungin64 µg/mLCheng et al., 2023 [64]
Enfumafungin B>64 µg/mL
Enfumafungin C>64 µg/mL
Sphaceloma sp. from the leaf of Poplar sp.Persephacin2.5 µg/mLDu et al., 2023 [65]
Myrothecium inundatumMyropeptin C16 µg/mLJagels et al., 2023 [66]
Myropeptin D16 µg/mL
Myropeptin E16 µg/mL
Myropeptin A14 µg/mL
Hypomyces pseudocorticiicola FKA-73Hakuhybotrol >128 µg/mLWatanabe et al., 2023 [67]
Cladobotric acids F>128 µg/mL
Pyrenulic acid A 16 µg/mL
F2928-1 2 µg/mL
Cladobotric acids E2 to 4 µg/mL
Cladobotric acids H16 to 32 µg/mL
Cladobotric acids A4 to 8 µg/mL
Lactobacillus paracasei 28.4Culture extract 3.75 to 7.5 mg/mLRossoni et al., 2020 [68]
Oregano or thyme and other aromatic plants. Carvacrol125–500 µg/mLIsmail et al., 2022 [69]
Medicinal plantsGeraniol225 µg/mLFatima et al., 2023 [70]
Different Streptomyces spp.Nikkomycin Z0.125 to >64 µg/mLBentz et al., 2021 [71]
Medicinal plants6-shogaol16–32 µg/mLKim & Eom, 2021 [72]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Ganeshkumar, A.; Muthuselvam, M.; Lima, P.M.N.d.; Rajaram, R.; Junqueira, J.C. Current Perspectives of Antifungal Therapy: A Special Focus on Candida auris. J. Fungi 2024, 10, 408. https://doi.org/10.3390/jof10060408

AMA Style

Ganeshkumar A, Muthuselvam M, Lima PMNd, Rajaram R, Junqueira JC. Current Perspectives of Antifungal Therapy: A Special Focus on Candida auris. Journal of Fungi. 2024; 10(6):408. https://doi.org/10.3390/jof10060408

Chicago/Turabian Style

Ganeshkumar, Arumugam, Manickam Muthuselvam, Patricia Michelle Nagai de Lima, Rajendren Rajaram, and Juliana Campos Junqueira. 2024. "Current Perspectives of Antifungal Therapy: A Special Focus on Candida auris" Journal of Fungi 10, no. 6: 408. https://doi.org/10.3390/jof10060408

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop