Next Article in Journal
Microbial Resource Management for Ex Situ Biomethanation of Hydrogen at Alkaline pH
Next Article in Special Issue
Indoor Dust as a Source of Virulent Strains of the Agents of Cryptococcosis in the Rio Negro Micro-Region of the Brazilian Amazon
Previous Article in Journal
Glycosphingolipids Recognized by Acinetobacter baumannii
Previous Article in Special Issue
Environmental Factors That Contribute to the Maintenance of Cryptococcus neoformans Pathogenesis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

New Approaches for Cryptococcosis Treatment

1
Laboratory of Antifungal Chemotherapy, Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil
2
Laboratory of Nanomedicine and Drug Delivery Systems, Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil
*
Author to whom correspondence should be addressed.
Microorganisms 2020, 8(4), 613; https://doi.org/10.3390/microorganisms8040613
Submission received: 26 February 2020 / Revised: 16 April 2020 / Accepted: 17 April 2020 / Published: 23 April 2020
(This article belongs to the Special Issue Current Understanding of Cryptococcus and Cryptococcosis)

Abstract

:
Cryptococcosis is an important opportunistic infection and a leading cause of meningitis in patients with HIV infection. The antifungal pharmacological treatment is limited to amphotericin B, fluconazole and 5- flucytosine. In addition to the limited pharmacological options, the high toxicity, increased resistance rate and difficulty of the currently available antifungal molecules to cross the blood–brain barrier hamper the treatment. Thus, the search for new alternatives for the treatment of cryptococcal meningitis is extremely necessary. In this review, we describe the therapeutic strategies currently available, discuss new molecules with antifungal potential in different phases of clinical trials and in advanced pre-clinical phase, and examine drug nanocarriers to improve delivery to the central nervous system.

1. Introduction

Cryptococcus infection and development of cryptococcosis in humans generally occurs due to inhalation of yeasts or spores of Cryptococcus neoformans and Cryptococcus gattii present in the environment [1,2]. Cryptococcus causes pulmonary cryptococcosis, and in some patients, the infection may remain latent or oligosymptomatic for a long period. After pulmonary infection, most cases evolve to hematogenous dissemination, with a special predilection for the central nervous system (CNS) leading to cryptococcal meningitis (~90%); in addition, the occurrence of lesions in other tissues is a serious sign of fungus spread [1,3,4].
As a common opportunistic infection in patients with advanced HIV infection, cryptococcosis is the leading cause of meningitis accounting for ~223,100 cases/year, and over 81% mortality in the world [5]. Cryptococcosis-related deaths are most frequent in the sub-Saharan Africa, where mortality reaches 70% [6]. Although access to antiretroviral therapy has resulted in a substantial reduction in the incidence of meningitis by Cryptococcus in high-income countries, this infection is likely to remain a major cause of HIV-related mortality in low- and middle-income countries, where antiretroviral treatment is insufficient/unavailable and begins at an advanced stage of HIV/AIDS [7,8].
The antifungal treatment depends of the cryptococcosis clinical form and immunological state of the patient [9,10]. The current antifungal arsenal available for cryptococcosis treatment is limited to three drugs, used alone or in combination: Amphotericin B deoxycholate (AMB) and its lipid formulations (liposomal AMB (LAMB), AMB lipid complex (ABLC), and AMB colloidal dispersion (ABCD)), flucytosine (5-fluorocytosine or 5-FC), and fluconazole (FLC) [9]. In addition to the limited therapeutic options, high attendance and recurrence rates due to the increased resistance of Cryptococcus to FLC and 5-FC have been reported [11,12].
Treatment of CNS infections is often difficult because the blood–brain barrier (BBB) limits the diffusion of molecules to the brain tissues, and efflux pumps reduce drug concentrations in the CNS [13]. To gain access to the CNS, drugs can also pass through tight junctions that are much smaller in the BBB (20 Å) than in other tissues of the organism (100 Å), which prevent the diffusion of drugs with high molecular weight (MW). The upper MW limit for efficient crossing of BBB is 400–500 g/mol [14,15], and beyond that, higher lipophilicity and volume of distribution are important properties associated with maximal CNS exposure [16].
Among the antifungals available for the treatment of cryptococcal meningitis, 5-FC (MW = 120 g/mol) and FLC (MW = 309 g/mol) diffuse more easily and present excellent cerebrospinal fluid (CSF) and brain tissue penetration (52–100% of serum concentration) [14,16]. In contrast, AMB is composed of large molecules (MW = 924 g/mol), and although AMB deoxycholate and lipid formulations (ABLC and LAMB) have been previously associated with low penetration in the CSF and brain, the antifungal therapy with these formulations resulted in clinical success [16]. Interestingly, LAMB showed lower penetration in the brain tissue than the AMB deoxycholate formulation (3% vs. 27%) [16].
This scenario emphasizes the pressing need for new strategies and alternatives for the antifungal treatment of cryptococcosis, especially the meningitis. In this review, we describe the conventional therapy of cryptococcosis and the main characteristics of the antifungals currently used; and we discuss new antifungal molecules with anti-Cryptococcus activity potential and nanocarrier-based formulations to aid antifungals penetration in the CNS.

2. Current Therapy

The treatment of cryptococcal meningitis consists of three phases: induction (2 weeks), consolidation (8 weeks) and maintenance (6–12 months). The guidelines of the Society for Infectious Diseases of America [9] and the World Health Organization [17] emphasize the importance of the use of potent fungicidal drugs during the induction phase; however, worldwide access to antifungal drugs is still inadequate [18], which highlights the importance of alternative treatment strategies.
The primary therapy of cryptococcal meningitis depends on the condition of the patients infected with Cryptococcus. For HIV-infected, HIV-non infected and non-transplanted individuals, the primary therapy consists on the induction with AMB (0.7–1.0 mg/kg/day) plus 5-FC (100 mg/kg/day) for 2 weeks. For consolidation and maintenance, FLC at 400 mg/day for 8 weeks (minimum) and at 200 mg/day for 6–12 months, respectively, are employed. In addition, there are other alternative regimens; for example, in case of AMB intolerance, LAMB (3–4 mg/kg/day) or ABLC (5 mg/kg/day) can be used. If 5-FC is not used, AMB deoxycholate or AMB lipid formulations should be maintained for at least 2 weeks [9].
For patients with nonmeningeal cryptococcosis forms as pulmonary (immunosuppressed and nonimmunosupressed) and nonpulmonary cryptococcosis, FLC (400 mg/day) for 6–12 months is recommended. For the pulmonary (nonimmunosupressed) form, voriconazole (VRC) (200 mg twice/day), itraconazole (ITC) (200 mg/day), and posaconazole (POS) (400 mg twice/day) are acceptable alternatives if FLC is unavailable or contraindicated [9].

2.1. Amphotericin B (AMB)

Despite AMB dose-limiting toxicity, it has remained the gold standard for treating disseminated life-threatening fungal infections [19]. Its fungicidal effect is associated with AMB binding to ergosterol in the membranes of fungal cell (Figure 1) [20,21]. AMB perturbs the membrane function, causing leakage of cellular contents, and leads to death by cellular dysfunction [20,21]. The first commercially available formulation was Fungizone®, a conventional micellar form of AMB and deoxycholate. Currently, parenteral formulations based on lipid carriers are also available, and they include a liposomal formulation (LAMB), a lipid complex formulation (ABLC), and a colloidal dispersion (ABCD) [22]. Their main advantage is the reduction of side effects of AMB [23].
Resistance to AMB is rare and often caused by a decrease in the amount of ergosterol in the plasma membrane or a change in the target sterol, which leads to a decrease in the binding of AMB [19,24]. Some fungal cells have a mutation in the ergosterol biosynthesis pathway, producing ergosterol-like compounds instead of ergosterol, which have lower binding affinity for AMB [25,26].

2.2. Flucytosine (5-FC)

5-FC was synthesized in 1957 as a potent antitumoral agent [27,28]. 5-FC is taken into the fungal cell by cytosine permease, and its action as an antifungal agent depends on its conversion to 5-fluorouracil (5-FU) within of the target cells. 5-FU becomes incorporated to the RNA and inhibits DNA synthesis by thymidylate synthase inhibition (Figure 1). It is most active agent against yeasts, including Candida and Cryptococcus spp. [28]; however, the occurrence of resistance to 5-FC prevents its use as a single agent [28,29,30,31,32]. Currently, its use is indicated only in combination with other antifungals, mainly AMB [23,28]. 5-FC exhibits significant adverse effects, in particular hepatotoxicity and myelotoxicity, which is probably due to toxic 5-FU plasma concentrations.

2.3. Fluconazole (FLC)

FLC is a triazole agent that inhibits the fungal cytochrome P450-dependent lanosterol C14-alpha-demethylase (Erg11 or Cyp51) leading to ergosterol biosynthesis inhibition (Figure 1) [12,33,34]. FLC diffuses easily into the cerebrospinal fluid, sputum and saliva, and is concentrated in the urine and skin [35]. The most frequent adverse effects are gastrointestinal events, headache and skin rashes; isolated instances of clinically evident hepatic dysfunction have occurred in patients with AIDS [36]. Over the years, there has been a gradual increase of resistance to FLC in clinical isolates of C. neoformans, and nowadays, resistance is a relatively common event in relapse episodes of cryptococcal meningitis [12]. FLC resistance phenotype in Cryptococcus spp. have been associated with mutations in the ERG11 gene [12,37,38]. However, heteroresistance in Cryptococcus spp. can lead to higher FLC tolerance by selection of heteroresistant clones after induction due to previous exposure to FLC [12,39].

2.4. Voriconazole (VRC)

VRC was developed to increase the antifungal spectrum of available triazoles. To reach this goal, the molecule of FLC was modified, with the substitution of the fluoropyrimidine ring for one of the azole groups, and addition of the α-methyl group to provide fungicidal activity against molds [40,41]. The most frequently reported adverse effect of VRC is transient visual disturbances, that are often associated with higher doses, and considerable hepatotoxic effects. In addition, studies reported important drug interactions with VRC [41]. An ongoing clinical trial study (from 2020 to 2022), named “Three Induction Treatments on Cryptococcal Meningitis (TITOC)”, is investigating its use for cryptococcosis treatment at the Hospital of the University of Zhejiang, China (NCT04072640, www.clinicaltrials.gov). Resistance to this azole is not a common event, but there are reports in the literature in recent years [42,43,44,45].

3. New Molecules and Drug Repurposing

Immunobiological and new molecules acting on non-conventional targets or other structures of the fungal cell might have potential as antifungal agents. In this context, drug repositioning is an interesting strategy for antifungal discovery because pharmacokinetics and safety data in humans have been previously established. Therefore, expanding the application of a drug to additional diseases is both cost and time-effective [46,47]. In this section, we will discuss new molecules and drugs currently used in the treatment of other diseases that have activity against Cryptococcus spp. (Figure 1).

3.1. Interferon-Gamma (IFN-γ)

IFN-γ is an endogenous cytokine with several biological properties and activities, including a key role in the host response to intracellular pathogens, directing the immune system towards the protective Th1 type immunity [48]. Exogenous IFN-γ has been investigated as a potential adjunct agent for treatment of cryptococcal meningitis. In a murine model of pulmonary and disseminated infection, IFN-γ administration resulted in the decrease of the fungal burden in the infected organs, and significantly extended mice survival [49]. One phase II clinical trial (NCT00012467) suggested that IFN-γ may provide rapid and early sterilization of CNS in patients with HIV-associated cryptococcal meningitis without pronounced adverse effects [48]. However, in another study, it was observed that even though administration of IFN-γ improved the fungal clearance from the CNS, it failed to significantly decrease patient mortality [50].

3.2. Mycograb

Mycograb is a recombinant human antibody against fungal heat shock protein 90 (Hsp90), which are chaperones required for the maintenance of cellular homeostasis in various fungal pathogens [51,52]. Cryptococcus neoformans isolates were susceptible to mycograb at MIC values from 256 to 1024 µg/mL, and a synergistic effect was observed in combination with AMB [53]. The efficacy and safety of mycograb for cryptococcal meningitis are under evaluation in ongoing phase II clinical trials (NCT00324025 and NCT00847678).

3.3. 18B7

18B7 is a monoclonal antibody directed against the capsular polysaccharide of C. neoformans. Administration of 18B7 promoted rapid clearance of serum cryptococcal antigen and deposition in the liver and spleen, and presented no reactivity with normal mouse, rat, or human tissues [54]. It also reduced the fungal burden in tissues, improved granuloma formation, and demonstrated synergism with AMB, FLC and 5-FC in mice [55,56,57,58]. 18B7 was evaluated in a phase I clinical trial in HIV-infected patients with cryptococcal meningitis, being well tolerated in doses up to 1 mg/kg without evidence of toxicity [59].

3.4. APX001 (Fosmanogepix)/APX001A (Manogepix)

APX001 (prodrug of APX001A) is a first-in-class small-molecule antifungal drug candidate that inhibits the fungal enzyme Gwt1 (an inositol acylase) in the glycosylphosphatidylinositol (GPI) biosynthesis pathway [60]. The APX001A MIC ranged from 0.03 to 2 µg/mL for 48 Cryptococcus spp. clinical isolates in vitro [61,62], and demonstrated in vitro synergism with FLC [62]. APX001 alone or in combination with FLC decreased the fungal burden in the lungs and brain using cryptococcal meningitis murine model [62]. Other structural analogues of APX001A also demonstrated an excellent in vitro inhibitory effect on C. neoformans; and using in vivo assay APX2096 (prodrug of APX2039) led to a nearly complete or complete sterilization of lungs and brain [62]. The preclinical efficacy of APX001/APX001A against Cryptococcus associated with previous safety and pharmacological data (NCT02957929 and NCT02956499) lend support to further clinical evaluation of the molecule for treatment of human cryptococcosis.

3.5. T-2307

T-2307 is a novel arylamidine derivative with broad-spectrum of action and potent in vitro and in vivo activities, that acts by selectively disrupting mitochondrial function in yeasts [63]. The antifungal activity for C. neoformans was observed at MIC ranging from 0.0039 to 0.0625 µg/mL [64], and for C. gattii at 0.0078–0.0625 µg/mL [65]. The efficacy of T-2307 was confirmed in murine models of cryptococcosis: at 0.1 mg/kg, T-2307 significantly delayed mortality in mice infected by C. neoformans when compared with the untreated group, and T-2307 exhibited a superior protective effect compared to AMB at similar treatment regimens [64]. Administration of T-2307 alone at 2 mg/kg/day significantly reduced viable cell counts in the lungs and brain of mice infected by C. gattii and the results were similar to standard treatments [65].

3.6. Sertraline

Sertraline is an antidepressant that belongs to the group of selective inhibitors of serotonin reuptake. Initially used for treatment of major depressive disorder, it is now also approved for management of obsessive-compulsive, panic and post-traumatic stress disorders [66]. Although its mechanism of action on fungi has not fully elucidated, inhibition of protein synthesis in Cryptococcus spp. has been described [67]. In vitro studies showed that sertraline is effective to inhibit Cryptococcus growth at 1–8 µg/mL; in contrast to FLC, sertraline showed fungicidal effect at concentrations higher than 6 µg/mL [67,68]. Murine cryptococcosis model confirmed the antifungal activity observed in vitro, in which sertraline at 15 mg/kg decreased the fungal burden in the brain and spleen when compared with the untreated group [67,68]. Sertraline combined with FLC in vitro showed either additive or synergistic effects, and in animal models, this drug combination led to fungal clearance at a greater rate than either drug alone [67,69,70]. Sertraline use for cryptococcal meningitis treatment alone or in combination with AMB and FLC was investigated in phase III clinical trials (NCT01802385 and NCT03002012), and these studies demonstrated that sertraline did not reduce the mortality rate of patients. This lack of efficacy appears to be multifactorial, and might be associated with insufficient duration of therapeutic sertraline concentrations [71].

3.7. Tamoxifen

Tamoxifen belongs to the pharmacological class of selective estrogen receptor modulators; it is an estrogen receptor agonist in the bone, cardiovascular system, and endometrium, while acting as an estrogen receptor antagonist in the breast tissue. This drug is clinically used to treat and prevent breast cancer and osteoporosis [72]. Tamoxifen has in vitro antifungal activity against Cryptococcus spp. clinical isolates, with MIC ranging from 2 to 16 µg/mL, acting synergistically when combined with AMB and FLC [73,74]. In the murine disseminated cryptococcosis model, treatment with tamoxifen at 200 mg/kg/day combined with FLC at 5 mg/kg/day decreased the burden fungal by ~1 log in the brain tissue [74]. The authors of the study suggested the use of this drug for treatment of cryptococcosis because high concentrations (well above of the MIC values) were reached in the CNS in addition to the antifungal activity inside macrophages, synergism with existing therapies AMB and FLC, and good oral bioavailability [72,74]. At the moment, clinical trials (phase II) are being carried out to evaluate the efficacy, feasibility, and safety of tamoxifen in combination with standard therapies (AMB and FLC) in the treatment of cryptococcal meningitis (NCT03112031). Although tamoxifen activity against Cryptococcus has been reported, and the drug is under evaluation in ongoing clinical trials for cryptococcosis treatment, its mechanism of action has not been elucidated yet.

3.8. AR-12

AR-12, a small molecule derived from celecoxib, was tested as an antitumoral agent in phase I clinical trials, and licensed to Arno Therapeutics (NCT00978523) [75]. AR-12 is a non-nucleoside acetyl CoA synthetase inhibitor as previously investigated in S. cerevisiae and C. albicans [76]. This molecule has broad-spectrum antifungal activity, including for C. neoformans, with MIC value of 4 µg/mL, and AR-12 was demonstrated to be effective in a murine model of disseminated cryptococcosis when combined with FLC (dose at 100 and 10 mg/kg, respectively), decreasing the fungal burden in the brain [77].

3.9. Miltefosine (MFS)

MFS belongs to the alkylphosphocholine class of molecules, and is used in the treatment of cutaneous metastases of breast cancer and leishmaniasis [78]. Studies showed that MFS has a broad-spectrum in vitro antifungal activity, including against C. gattii and C. neoformans isolates in the both planktonic (0.25–4 µg/mL) and biofilm (8 - ≥16 µg/mL) lifestyles [79,80,81]. Moreover, MFS was effective to control the fungal infection in the larval model of Galleria mellonella by C. gattii at 10, 20, and 40 mg/kg [82]. MFS at 3.6 and 7.2 mg/kg/day has shown effectiveness in the murine model of disseminated cryptococcosis [80] although this result has been conflicting with other work [83]. This contradiction demonstrates variable translation of in vitro MFS activity to in vivo murine models of disseminated cryptococcosis. Studies evidenced that MFS acts through multiple mechanisms, being able to alter membrane permeability, inhibit phospholipase B1 and induce an apoptotic-like cell death reducing the mitochondrial membrane potential, increasing reactive oxygen species (ROS) production, and inducing DNA fragmentation and condensation [80,81]. Despite the extensive and exciting in vitro reports highlighting MFS usefulness as an antifungal drug, no clinical trial for treatment of fungal infections is under way.

3.10. Tetrazoles

Tetrazoles are synthetic molecules produced from azoles, non-metabolized bioisosteric analogs of carboxylic acid and cis-amide; they possess diverse chemotherapeutic properties and are highly selective fungal Cyp51 inhibitor [84,85]. Among tetrazoles, VT-1129 and VT-1598 are more selective for fungal Cyp51 than mammalian Cyp450 enzymes and both molecules showed antifungal efficacy against Cryptococcus spp. [86,87]. VT-1129 inhibited the growth of C. neoformans and C. gattii isolates at 0.003–4 µg/mL and 0.06–8 µg/mL, respectively [88,89]. VT-1598 has lower MIC values (0.06 to 0.15 µg/mL) [90,91]. In addition to the in vitro models, assays using cryptococcosis murine models demonstrated that oral administration of VT-1598 resulted in suitable plasma and brain concentrations, leading to a significant reduction in the brain fungal burden [91]. Recently, phase I clinical trials have started to assess the safety and pharmacokinetics of VT-1598 (NCT04208321).

4. Other Molecules and Targets

To find alternatives for cryptococcosis treatment, other molecules have been evaluated in the pre-clinical studies. In vitro screening with some off-patent drugs found 43 drugs capable of inhibiting the growth of C. neoformans, such as cliclopirox and auranofin [92]. Other studies have investigated the use of antiprotozoal drugs as benzimidazoles with MIC ranging from 0.125 to 0.45 µg/mL, and flubendazole was found to reduce fungal burden in infected mice [93,94].
Drugs with action on Cryptococcus virulence mechanisms have the potential to aid treatment. For example, the herbicide glyphosate inhibited melanization of yeasts at subinhibitory concentrations in vitro, and its administration in mice infected with C. neoformans delayed the melanization of yeasts and prolonged mice survival twice as long compared to untreated mice [95]. The vesicular transport system is essential for virulence, being related to the assembly of the mucopolysaccharide capsule, secretion of melanin, lipids, hydrolytic enzymes and other molecules [96]. Two synthetic compounds, N-(3-bromo-4-hydroxybenzylidene)-2-methylbenzohydrazide (BHBM) and 3-bromo-N-(3-bromo-4-hydroxybenzylidene) benzohydrazide (DO), caused accumulation of intracellular vesicles and inhibited surface glucosylceramides, demonstrating antifungal potential for Cryptococcus [97].

5. Drug Delivery Systems

Drug delivery systems represent a promising alternative for treatment of diseases that affect the brain due to the possibility of enabling drug transport across BBB. They can improve transport by masking physicochemical characteristics of drugs, which circumvents the need for molecular modifications [98]. In addition, these carriers are small in size and can be composed of various materials, such as polymers, lipids, metals and inorganic elements, among others. The most frequently used nanocarriers include liposomes, micelles, polymeric and inorganic nanoparticles, such as gold and silver (Figure 2A) [99,100]. Liposomes are biodegradable colloidal aggregates composed of one or multiple bilayers that encompass an internal aqueous compartment [100]. Nanoparticles are colloidal systems with a compact structure that vary in size from 10 to 1000 nm, in which the drug can be dissolved, dispersed, trapped, encapsulated or attached to a matrix; they can be produced with inorganic or organic materials as PLGA, PLA, PBCA, albumin, among others [101,102].
Nanocarriers may undergo surface modifications to improve drug penetration into the brain, which might involve various processes as represented in Figure 2B,C. Nanocarrier surface modification enables recognition/targeting of specific ligands on the cell surface, leading to cell uptake and transport into the brain after systemic administration via receptor-mediated pathways [103,104]. For example, cationic nanocarriers can interact electrostatically with BBB (which has a net negative charge), and be internalized by adsorption-mediated endocytosis [101]. Other mechanisms that might participate in the ability of nanocarriers to improve transport across BBB include (i) a general surfactant effect, characterized by solubilization and fluidization of the cell membrane, (ii) ability to open tight junctions, and (iii) inhibition of efflux transporters (such as P-glycoprotein) at the BBB, which is largely attributed to the presence of specific surfactants at the nanocarrier surface, such as polysorbate 80 and vitamin E TPGS [105,106]. In addition to polysorbate 80, other compounds have been previously employed to modify the surface of nanocarriers intended for encapsulation of antifungals and treatment of cryptococcal meningitis, including angiopep-2, apolipoprotein E and borneol. In this section, we will discuss nanocarriers with surface modifications aimed at improving transport of antifungal agents across BBB for treatment of cryptococcal meningitis.
AMB is considered the gold standard for cryptococcal meningitis treatment (induction phase) and for other systemic fungal infections despite its multiple adverse effects. Lipid-based nanocarriers (LAMB, ABDC, ABLC) have been shown to reduce AMB toxicity, but not to increase its penetration in the CNS when compared with the deoxycholate (conventional) AMB formulation [16]. This has motivated research for other nanocarriers. A new oral cochleate-amphotericin B formulation (CAMB or MAT2203) has been evaluated in a phase II clinical trial for efficacy and safety in the treatment of cryptococcosis (NCT03196921). Cochleates are generally described as stable phospholipid-cation crystalline structures consisting of a spiral lipid bilayer sheet with no internal aqueous space [107]. In a murine model of infection with C. neoformans, oral administration of CAMB delivered AMB at therapeutic levels to the CNS [108]. Moreover, its co-administration with 5-FC showed an equivalent efficacy to deoxycholate AMB with 5-FC, and superior to oral FLC, but with reduced toxicity [108].
Polymeric nanoparticles coated with polysorbate 80, a non-ionic surfactant, have been shown to increase transport through BBB and consequently, improve drug concentrations in the CNS. AMB-poly(lactic acid)-b-poly(ethylene glycol) (AMB-PLA-b-PEG) nanoparticles prepared by nanoprecipitation and coated with polysorbate 80 were able to increase AMB concentration in the brain, suggesting that polysorbate 80 aids transport across the BBB. The formulation significantly reduced the fungal burden in the brain of a mice model of cryptococcosis when compared with deoxycholate AMB, and decreased AMB toxicity [109]. Another type of nanoparticle, poly butyl cyanoacrylate nanoparticles (AMB-PBCA-NP) coated with polysorbate 80 were detected in the brain 30 min after systemic administration and had a higher brain tissue concentration than LAMB. In addition, the survival rate of mice treated with AMB-PBCA- NP (80%) was significantly higher than LAMB (60%) and deoxycholate AMB (0%) [110].
Shao et al. (2010) demonstrated that an angiopep-2 modified PE-PEG (1,2-Distearoyl-sn-glycero-3-phosphoethanolamine-N-[methoxy (polyethylene glycol)-2000]) based micellar drug delivery system loaded with AMB (Angiopep-PEG-PE/AMB) was more effectively transported across the BBB than the deoxycholate AMB and the micelle without angiopep-2 in in vitro and in vivo assays. This superiority was attributed to angiopep-2 being a ligand of low-density lipoprotein receptor-related protein (LRP) present in the brain, which has been reported to mediate the transport of ligands across endothelial cells of the BBB [111]. When Angiopep-PEG-PE/AMB and AMB commercial formulations were compared for treatment of murine cryptococcosis, Angiopep-PEG-PE/AMB was more effective, leading to the highest AMB concentration in the brain, reducing the fungal burden and prolonging the median survival time of mice [112].
ITC and its derivative compound POS, both triazole agents, are acceptable alternatives for treatment of the pulmonary form of cryptococcosis, but not for cryptococcal meningitis due to its difficulty of crossing the BBB [9]. ĆURIĆ and contributors developed poly(butyl cyanoacrylate) nanoparticles for ITC [113], and demonstrated that nanoparticles functionalization with covalent binding of apolipoprotein E enabled targeting of low-density lipoprotein receptor (LDLR) expressed on the endothelial brain capillary cell membrane [114]. ITC was also incorporated in bovine serum albumin nanoparticles (ITC-BSA-NP) modified with borneol (BO) and polyethylene glycol (PEG) (PEG/BO-ITC-BSA-NP) [115]. The nanoparticles significantly increased ITC uptake by the bEnd.3 cells (a mouse brain cell line) and promoted ~2-fold higher brain uptake in mice than ITC conventional formulation [115]. Borneol is a highly lipid-soluble bicyclic monoterpene that can facilitate penetration of drugs in the brain due to BBB opening [116]. Another study used a 29-amino-acid peptide derived from rabies virus glycoprotein (RVG29) conjugated with albumin nanoparticles as carrier for ITC (RVG29-ITC-NP), and demonstrated a significant drug accumulation in the brain compared to albumin nanoparticles without RVG29 and a cyclodextrin formulation of ITC [117].

6. Concluding Remarks

Searching for new alternatives for treatment of fungal infections has been pivotal. Here, we highlighted the remarkable deficit of antifungal options in the treatment of cryptococcosis recommended by the current guidelines. Studies have been carried out with the intention of finding new therapeutic alternatives as described in this work; some of these molecules and the use of nanocarriers to improve drug delivery into the CNS may become future therapies for cryptococcal meningitis.

Author Contributions

All authors contributed to drafting or revising the manuscript, gave final approval of the version to be published, and agreed to be accountable for all aspects of the work. All authors have read and agreed to the published version of the manuscript.

Funding

This study was supported by Fundação de Amparo à Pesquisa do Estado de São Paulo (FAPESP, grant numbers 2017/19374-9 and 2018/13877-1). CCS was a FAPESP fellow (2018/12149-2) and KI is a research fellow of the Conselho Nacional de Desenvolvimento Científico e Tecnológico (CNPq, grant 303373/2019-9).

Acknowledgments

We are grateful to Eduardo A. M. Nogueira for the watercolor art of Figure 1.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Maziarz, E.K.; Perfect, J.R. Cryptococcosis. Infect. Dis. Clin. N. Am. 2016, 30, 179–206. [Google Scholar] [CrossRef] [Green Version]
  2. May, R.C.; Stone, N.R.H.; Wiesner, D.L.; Bicanic, T.; Nielsen, K. Cryptococcus: From environmental saprophyte to global pathogen. Nat. Rev. Microbiol. 2016, 14, 106–117. [Google Scholar] [CrossRef]
  3. Lin, X.; Heitman, J. The biology of the Cryptococcus neoformans species complex. Annu. Rev. Microbiol. 2006, 60, 69–105. [Google Scholar] [CrossRef] [PubMed]
  4. O’Meara, T.R.; Andrew Alspaugh, J. The Cryptococcus neoformans capsule: A sword and a shield. Clin. Microbiol. Rev. 2012, 25, 387–408. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Rajasingham, R.; Smith, R.M.; Park, B.J.; Jarvis, J.N.; Govender, N.P.; Chiller, T.M.; Denning, D.W.; Loyse, A.; Boulware, D.R. Global burden of disease of HIV-associated cryptococcal meningitis: An updated analysis. Lancet Infect. Dis. 2017, 17, 873–881. [Google Scholar] [CrossRef] [Green Version]
  6. Limper, A.H.; Adenis, A.; Le, T.; Harrison, T.S. Fungal infections in HIV/AIDS. Lancet Infect. Dis. 2017, 3099, 1–10. [Google Scholar] [CrossRef]
  7. Loyse, A.; Thangaraj, H.; Easterbrook, P.; Ford, N.; Roy, M.; Chiller, T.; Govender, N.; Harrison, T.S.; Bicanic, T. Cryptococcal meningitis: Improving access to essential antifungal medicines in resource-poor countries. Lancet Infect. Dis. 2013, 13, 629–637. [Google Scholar] [CrossRef] [Green Version]
  8. Loyse, A.; Burry, J.; Cohn, J.; Ford, N.; Chiller, T.; Ribeiro, I.; Koulla-Shiro, S.; Mghamba, J.; Ramadhani, A.; Nyirenda, R.; et al. Leave no one behind: Response to new evidence and guidelines for the management of cryptococcal meningitis in low-income and middle-income countries. Lancet Infect. Dis. 2019, 19, e143–e147. [Google Scholar] [CrossRef]
  9. Perfect, J.R.; Dismukes, W.E.; Dromer, F.; Goldman, D.L.; Graybill, J.R.; Hamill, R.J.; Harrison, T.S.; Larsen, R.A.; Lortholary, O.; Nguyen, M.-H.; et al. Clinical practice guidelines for the management of cryptococcal disease: 2010 update by the infectious diseases society of america. Clin. Infect. Dis. 2010, 50, 291–322. [Google Scholar] [CrossRef] [Green Version]
  10. World Health Organization. Guidelines for the Diagnosis, Prevention, and Management of Cryptococcal Disease in HIV-Infected Adults, Adolescents and Children. 2019. Available online: https://www.who.int/hiv/pub/guidelines/cryptococcal-disease/en/ (accessed on 20 February 2019).
  11. Cheong, J.W.S.; Mccormack, J. Fluconazole resistance in cryptococcal disease: Emerging or intrinsic? Med. Mycol. 2013, 51, 261–269. [Google Scholar] [CrossRef] [Green Version]
  12. Bongomin, F.; Oladele, R.O.; Gago, S.; Moore, C.B.; Richardson, M.D. A systematic review of fluconazole resistance in clinical isolates of Cryptococcus species. Mycoses 2018, 61, 290–297. [Google Scholar] [CrossRef] [PubMed]
  13. Wirth, F.; de Azevedo, M.I.; Pilla, C.; Aquino, V.R.; Neto, G.W.; Goldani, L.Z. Relationship between intracranial pressure and antifungal agents levels in the CSF of patients with cryptococcal meningitis. Med. Mycol. 2018, 56, 257–262. [Google Scholar] [CrossRef] [PubMed]
  14. Kethireddy, S.; Andes, D. CNS pharmacokinetics of antifungal agents. Expert Opin. Drug Metab. Toxicol. 2007, 3, 573–581. [Google Scholar] [CrossRef] [PubMed]
  15. Pardridge, W.M. Blood-brain barrier drug Targeting: The future of brain drug development. Mol. Interv. 2003, 3, 90–105. [Google Scholar] [CrossRef] [PubMed]
  16. Ashley, E.D. Antifungal Drugs: Special problems treating Central Nervous System infections. J. Fungi 2019, 5, 97. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. World Health Organization. Guidelines for the Diagnosis, Prevention and Management of Cryptococcal Disease in HIV-Infected Adults, Adolescents and Children: Supplement to the 2016 Consolidated Guidelines on the Use of Antiretroviral Drugs for Treating and Preventing HIV Infection; World Health Organization: Geneva, Switzerland, 2018; pp. 1–51. [Google Scholar]
  18. Perfect, J.R.; Bicanic, T. Cryptococcosis diagnosis and treatment: What do we know now. Fungal Genet. Biol. 2015, 78, 49–54. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  19. Ellis, D. Amphotericin B: Spectrum and resistance. J. Antimicrob. Chemother. 2002, 49, 7–10. [Google Scholar] [CrossRef] [Green Version]
  20. Wilcock, B.C.; Endo, M.M.; Uno, B.E.; Burke, M.D. C2′-OH of Amphotericin B plays an important role in binding the primary sterol of human cells but not yeast cells. J. Am. Chem. Soc. 2013, 135, 8488–8491. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  21. Liu, M.; Chen, M.; Yang, Z. Design of amphotericin B oral formulation for antifungal therapy. Drug Deliv. 2017, 24, 1–9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Cuddihy, G.; Wasan, E.K.; Di, Y.; Wasan, K.M. The development of oral amphotericin B to treat systemic fungal and parasitic infections: Has the myth been finally realized? Pharmaceutics 2019, 11, 99. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Bellmann, R.; Smuszkiewicz, P. Pharmacokinetics of antifungal drugs: Practical implications for optimized treatment of patients. Infection 2017, 45, 737–779. [Google Scholar] [CrossRef] [PubMed]
  24. Lozano-Chiu, M.; Rex, J.H. Resistance to antifungal agents. In Topley & Wilson’s Microbiology and Microbial Infections, 9th ed.; Arnold: London, UK, 1998; pp. 177–187. [Google Scholar]
  25. Posch, W.; Blatzer, M.; Wilflingseder, D.; Lass-Flörl, C. Aspergillus terreus: Novel lessons learned on amphotericin B resistance. Med. Mycol. 2018, 56, S73–S82. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. O’Shaughnessy, E.M.; Lyman, C.A.; Walsh, T.J. Amphotericin B: Polyene resistance mechanisms. In Antimicrobial Drug Resistance; Humana Press: Totowa, NJ, USA, 2009; pp. 295–305. [Google Scholar]
  27. Duschinsky, R.; Pleven, E.; Heidelberger, C. The synthesis of 5-Fluoropyrimidines. J. Am. Chem. Soc. 1957, 79, 4559–4560. [Google Scholar] [CrossRef]
  28. Vermes, A.; Guchelaar, H.; Dankert, J. Flucytosine: A review of its pharmacology, clinical indications, pharmacokinetics, toxicity and drug interactions. J. Antimicrob. Chemother. 2000, 46, 171–179. [Google Scholar] [CrossRef]
  29. Tassel, D.; Madoff, M. Treatment of Candida sepsis and Cryptococcus meningitis with 5-Fluorocytosine. JAMA 1968, 206, 830. [Google Scholar] [CrossRef]
  30. Polak, A.; Scholer, H.J. Mode of action of 5-Fluorocytosine and mechanisms of resistance. Chemotherapy 1975, 21, 113–130. [Google Scholar] [CrossRef]
  31. Normark, S.; Schonebeck, J. In Vitro studies of 5-Fluorocytosine resistance in Candida albicans and Torulopsis glabrata. Antimicrob. Agents Chemother. 1972, 2, 114–121. [Google Scholar] [CrossRef] [Green Version]
  32. Pfaller, M.A. Antifungal drug resistance: Mechanisms, epidemiology, and consequences for treatment. Am. J. Med. 2012, 125, S3–S13. [Google Scholar] [CrossRef]
  33. Richardson, K. The discovery and profile of Fluconazole. J. Chemother. 1990, 2, 51–54. [Google Scholar] [CrossRef]
  34. Kartalija, M.; Kaye, K.; Tureen, J.H.; Liu, Q.; Tauber, M.G.; Elliott, B.R.; Sande, M.A. Treatment of experimental Cryptococcal meningitis with Fluconazole: Impact of dose and addition of Flucytosine on mycologic and pathophysiologic outcome. J. Infect. Dis. 1996, 173, 1216–1221. [Google Scholar] [CrossRef] [Green Version]
  35. Brammer, K.W.; Farrow, P.R.; Faulkner, J.K. Pharmacokinetics and tissue penetration of Fluconazole in humans. Clin. Infect. Dis. 1990, 12, S318–S326. [Google Scholar] [CrossRef] [PubMed]
  36. Goa, K.L.; Barradell, L.B. Fluconazole. Drugs 1995, 50, 658–690. [Google Scholar] [CrossRef] [PubMed]
  37. Gago, S.; Serrano, C.; Alastruey-Izquierdo, A.; Cuesta, I.; Martín-Mazuelos, E.; Aller, A.I.; Gómez-López, A.; Mellado, E. Molecular identification, antifungal resistance and virulence of Cryptococcus neoformans and Cryptococcus deneoformans isolated in Seville, Spain. Mycoses 2017, 60, 40–50. [Google Scholar] [CrossRef] [PubMed]
  38. Rodero, L.; Mellado, E.; Rodriguez, A.C.; Salve, A.; Guelfand, L.; Cahn, P.; Cuenca-Estrella, M.; Davel, G.; Rodriguez-Tudela, J.L. G484S amino acid substitution in Lanosterol 14-α Demethylase (ERG11) is related to Fluconazole resistance in a recurrent Cryptococcus neoformans clinical isolate. Antimicrob. Agents Chemother. 2003, 47, 3653–3656. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  39. Sionov, E.; Chang, Y.C.; Garraffo, H.M.; Kwon-Chung, K.J. Heteroresistance to Fluconazole in Cryptococcus neoformans is intrinsic and associated with virulence. Antimicrob. Agents Chemother. 2009, 53, 2804–2815. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  40. Boucher, H.W.; Groll, A.H.; Chiou, C.C.; Walsh, T.J. Newer systemic antifungal agents. Drugs 2004, 64, 1997–2020. [Google Scholar] [CrossRef]
  41. Theuretzbacher, U.; Ihle, F.; Derendorf, H. Pharmacokinetic/Pharmacodynamic profile of Voriconazole. Clin. Pharmacokinet. 2006, 45, 649–663. [Google Scholar] [CrossRef]
  42. Mondon, P.; Petter, R.; Amalfitano, G.; Luzzati, R.; Concia, E.; Polacheck, I.; Kwon-Chung, K.J. Heteroresistance to Fluconazole and Voriconazole in Cryptococcus neoformans. Antimicrob. Agents Chemother. 1999, 43, 1856–1861. [Google Scholar] [CrossRef] [Green Version]
  43. Mandras, N.; Roana, J.; Tullio, V.; Allizond, V.; Banche, G.; Scalas, D.; Fucale, G.; Cuffini, A. A case of Fluconazole, Voriconazole-resistant Cryptococcus neoformans isolated from an immunocompetent patient. J. Chemother. 2011, 23, 379–380. [Google Scholar] [CrossRef]
  44. Perkins, A.; Gomez-Lopez, A.; Mellado, E.; Rodriguez-Tudela, J.L.; Cuenca-Estrella, M. Rates of antifungal resistance among Spanish clinical isolates of Cryptococcus neoformans var. neoformans. J. Antimicrob. Chemother. 2005, 56, 1144–1147. [Google Scholar] [CrossRef]
  45. Kano, R.; Okubo, M.; Hasegawa, A.; Kamata, H. Multi-azole-resistant strains of Cryptococcus neoformans var. grubii isolated from a FLZ-resistant strain by culturing in medium containing voriconazole. Med. Mycol. 2017, 55, 877–882. [Google Scholar] [PubMed]
  46. Tsubamoto, H.; Ueda, T.; Inoue, K.; Sakata, K.; Shibahara, H.; Sonoda, T. Repurposing itraconazole as an anticancer agent. Oncol. Lett. 2017, 14, 1240–1246. [Google Scholar] [CrossRef] [Green Version]
  47. Truong, M.; Monahan, L.G.; Carter, D.A.; Charles, I.G. Repurposing drugs to fast-track therapeutic agents for the treatment of cryptococcosis. PeerJ 2018, 6, e4761. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. Pappas, P.G.; Bustamante, B.; Ticona, E.; Hamill, R.J.; Johnson, P.C.; Reboli, A.; Aberg, J.; Hasbun, R.; Hsu, H.H. Recombinant Interferon-γ1b as adjunctive therapy for AIDS-related acute Cryptococcal meningitis. J. Infect. Dis. 2004, 189, 2185–2191. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Kawakami, K.; Tohyama, M.; Teruya, K.; Kudeken, N.; Xie, Q.; Saito, A. Contribution of interferon-γ in protecting mice during pulmonary and disseminated infection with Cryptococcus neoformans. FEMS Immunol. Med. Microbiol. 1996, 13, 123–130. [Google Scholar]
  50. Jarvis, J.N.; Meintjes, G.; Rebe, K.; Williams, G.N.; Bicanic, T.; Williams, A.; Schutz, C.; Bekker, L.-G.; Wood, R.; Harrison, T.S. Adjunctive interferon-γ immunotherapy for the treatment of HIV-associated cryptococcal meningitis. AIDS 2012, 26, 1105–1113. [Google Scholar] [CrossRef] [Green Version]
  51. Matthews, R.C.; Rigg, G.; Hodgetts, S.; Carter, T.; Chapman, C.; Gregory, C.; Illidge, C.; Burnie, J. Preclinical assessment of the efficacy of Mycograb, a human recombinant antibody against fungal HSP90. Antimicrob. Agents Chemother. 2003, 47, 2208–2216. [Google Scholar] [CrossRef] [Green Version]
  52. de Aguiar Cordeiro, R.; de Jesus Evangelista, A.J.; Serpa, R.; de Farias Marques, F.J.; de Melo, C.V.S.; de Oliveira, J.S.; Sidrim, J.J.C. Inhibition of heat-shock protein 90 enhances the susceptibility to antifungals and reduces the virulence of Cryptococcus neoformans/Cryptococcus gattii species complex. Microbiology 2016, 162, 309–317. [Google Scholar] [PubMed]
  53. Nooney, L.; Matthews, R.C.; Burnie, J.P. Evaluation of Mycograb®, amphotericin B, caspofungin, and fluconazole in combination against Cryptococcus neoformans by checkerboard and time-kill methodologies. Diagn. Microbiol. Infect. Dis. 2005, 51, 19–29. [Google Scholar] [CrossRef]
  54. Casadevall, A.; Cleare, W.; Feldmesser, M.; Glatman-Freedman, A.; Goldman, D.L.; Kozel, T.R.; Lendvai, N.; Mukherjee, J.; Pirofski, L.A.; Rivera, J.; et al. Characterization of a murine monoclonal antibody to Cryptococcus neoformans polysaccharide that is a candidate for human therapeutic studies. Antimicrob. Agents Chemother. 1998, 42, 1437–1446. [Google Scholar] [CrossRef] [Green Version]
  55. Mukherjee, J.; Feldmesser, M.; Scharff, M.D.; Casadevall, A. Monoclonal antibodies to Cryptococcus neoformans glucuronoxylomannan enhance fluconazole efficacy. Antimicrob. Agents Chemother. 1995, 39, 1398–1405. [Google Scholar] [CrossRef] [Green Version]
  56. Feldmesser, M.; Mukherjee, J.; Casadevall, A. Combination of 5-flucytosine and capsule-binding monoclonal antibody in the treatment of murine Cryptococcus neoformans infections and in vitro. J. Antimicrob. Chemother. 1996, 37, 617–622. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Dromer, F.; Charreire, J. Improved amphotericin B activity by a monoclonal anti-Cryptococcus neoformans antibody: Study during murine Cryptococcosis and mechanisms of action. J. Infect. Dis. 1991, 163, 1114–1120. [Google Scholar] [CrossRef] [PubMed]
  58. Feldmesser, M.; Casadevall, A. Effect of serum IgG1 to Cryptococcus neoformans glucuronoxylomannan on murine pulmonary infection. J. Immunol. 1997, 158, 790–799. [Google Scholar] [PubMed]
  59. Larsen, R.A.; Pappas, P.G.; Perfect, J.; Aberg, J.A.; Casadevall, A.; Cloud, G.A.; James, R.; Filler, S.; Dismukes, W.E. Phase I evaluation of the safety and pharmacokinetics of murine-derived anticryptococcal antibody 18B7 in subjects with treated Cryptococcal meningitis. Antimicrob. Agents Chemother. 2005, 49, 952–958. [Google Scholar] [CrossRef] [Green Version]
  60. Zhao, M.; Lepak, A.J.; Marchillo, K.; Vanhecker, J.; Sanchez, H.; Ambrose, P.G.; Andes, D.R. APX001 Pharmacokinetic/Pharmacodynamic target determination against Aspergillus fumigatus in an in vivo model of invasive pulmonary aspergillosis. Antimicrob. Agents Chemother. 2019, 63, e02372-18. [Google Scholar] [CrossRef] [Green Version]
  61. Pfaller, M.A.; Huband, M.D.; Flamm, R.K.; Bien, P.A.; Castanheira, M. In Vitro activity of APX001A (Manogepix) and comparator agents against 1,706 fungal isolates collected during an International Surveillance Program (2017). Antimicrob. Agents Chemother. 2019, 63, e00840-19. [Google Scholar] [CrossRef] [Green Version]
  62. Shaw, K.J.; Schell, W.A.; Covel, J.; Duboc, G.; Giamberardino, C.; Kapoor, M.; Moloney, M.; Soltow, Q.A.; Tenor, J.L.; Toffaletti, D.L.; et al. In Vitro and In Vivo evaluation of APX001A/APX001 and other Gwt1 inhibitors against Cryptococcus. Antimicrob. Agents Chemother. 2018, 62, e00523-18. [Google Scholar] [CrossRef] [Green Version]
  63. Shibata, T.; Takahashi, T.; Yamada, E.; Kimura, A.; Nishikawa, H.; Hayakawa, H.; Nomura, N.; Mitsuyama, J. T-2307 causes collapse of mitochondrial membrane potential in yeast. Antimicrob. Agents Chemother. 2012, 56, 5892–5897. [Google Scholar] [CrossRef] [Green Version]
  64. Mitsuyama, J.; Nomura, N.; Hashimoto, K.; Yamada, E.; Nishikawa, H.; Kaeriyama, M.; Kimura, A.; Todo, Y.; Narita, H. In Vitro and In Vivo antifungal activities of T-2307, a novel arylamidine. Antimicrob. Agents Chemother. 2008, 52, 1318–1324. [Google Scholar] [CrossRef] [Green Version]
  65. Nishikawa, H.; Fukuda, Y.; Mitsuyama, J.; Tashiro, M.; Tanaka, A.; Takazono, T.; Saijo, T.; Yamamoto, K.; Nakamura, S.; Imamura, Y.; et al. In vitro and in vivo antifungal activities of T-2307, a novel arylamidine, against Cryptococcus gattii: An emerging fungal pathogen. J. Antimicrob. Chemother. 2017, 72, 1709–1713. [Google Scholar] [CrossRef] [Green Version]
  66. DeVane, C.L.; Liston, H.L.; Markowitz, J.S. Clinical pharmacokinetics of Sertraline. Clin. Pharmacokinet. 2002, 41, 1247–1266. [Google Scholar] [CrossRef] [PubMed]
  67. Zhai, B.; Wu, C.; Wang, L.; Sachs, M.S.; Lin, X. The Antidepressant Sertraline provides a promising therapeutic option for neurotropic Cryptococcal infections. Antimicrob. Agents Chemother. 2012, 56, 3758–3766. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Trevinõ-Rangel, R.D.J.; Villanueva-Lozano, H.; Hernández-Rodríguez, P.; Martínez-Reséndez, M.F.; Garciá-Juárez, J.; Rodríguez-Rocha, H.; González, G.M. Activity of sertraline against Cryptococcus neoformans: In vitro and in vivo assays. Med. Mycol. 2016, 54, 280–286. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  69. Nayak, R.; Xu, J. Effects of sertraline hydrochloride and fluconazole combinations on Cryptococcus neoformans and Cryptococcus gattii. Mycology 2010, 1, 99–105. [Google Scholar] [CrossRef]
  70. Spitzer, M.; Griffiths, E.; Blakely, K.M.; Wildenhain, J.; Ejim, L.; Rossi, L.; De Pascale, G.; Curak, J.; Brown, E.; Tyers, M.; et al. Cross-species discovery of syncretic drug combinations that potentiate the antifungal fluconazole. Mol. Syst. Biol. 2011, 7, 1–14. [Google Scholar] [CrossRef] [Green Version]
  71. Rhein, J.; Huppler Hullsiek, K.; Tugume, L.; Nuwagira, E.; Mpoza, E.; Evans, E.E.; Kiggundu, R.; Pastick, K.A.; Ssebambulidde, K.; Akampurira, A.; et al. Adjunctive sertraline for HIV-associated cryptococcal meningitis: A randomised, placebo-controlled, double-blind phase 3 trial. Lancet Infect. Dis. 2019, 19, 843–851. [Google Scholar] [CrossRef]
  72. Morello, K.C.; Wurz, G.T.; DeGregorio, M.W. Pharmacokinetics of selective estrogen receptor modulators. Clin. Pharmacokinet. 2003, 42, 361–372. [Google Scholar] [CrossRef]
  73. Hai, T.P.; Van, A.D.; Ngan, N.T.T.; Nhat, L.T.H.; Lan, N.P.H.; Van Vinh Chau, N.; Thwaites, G.E.; Krysan, D.; Day, J.N. The combination of tamoxifen with amphotericin B, but not with fluconazole, has synergistic activity against the majority of clinical isolates of Cryptococcus neoformans. Mycoses 2019, 62, 818–825. [Google Scholar] [CrossRef] [Green Version]
  74. Butts, A.; Koselny, K.; Chabrier-Roselló, Y.; Semighini, C.P.; Brown, J.C.S.; Wang, X.; Annadurai, S.; DiDone, L.; Tabroff, J.; Childers, W.E.; et al. Estrogen receptor antagonists are anti-Cryptococcal agents that directly bind EF Hand proteins and synergize with Fluconazole In Vivo. MBio 2014, 5, e00765-13. [Google Scholar] [CrossRef] [Green Version]
  75. Santos-Gandelman, J.; Rodrigues, M.L.; Machado Silva, A. Future perspectives for cryptococcosis treatment. Expert Opin. Ther. Pat. 2018, 28, 625–634. [Google Scholar] [CrossRef] [PubMed]
  76. Koselny, K.; Green, J.; Favazzo, L.; Glazier, V.E.; DiDone, L.; Ransford, S.; Krysan, D.J. Antitumor/Antifungal celecoxib derivative AR-12 is a non-nucleoside inhibitor of the ANL-family adenylating enzyme Acetyl CoA synthetase. ACS Infect. Dis. 2016, 2, 268–280. [Google Scholar] [CrossRef] [PubMed]
  77. Koselny, K.; Green, J.; DiDone, L.; Halterman, J.P.; Fothergill, A.W.; Wiederhold, N.P.; Patterson, T.F.; Cushion, M.T.; Rappelye, C.; Wellington, M.; et al. The celecoxib derivative AR-12 has broad-spectrum antifungal activity in vitro and improves the activity of fluconazole in a murine model of cryptococcosis. Antimicrob. Agents Chemother. 2016, 60, 7115–7127. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. Sundar, S.; Chakravarty, J. An update on pharmacotherapy for leishmaniasis. Expert Opin. Pharmacother. 2015, 16, 237–252. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  79. Ravu, R.R.; Chen, Y.L.; Jacob, M.R.; Pan, X.; Agarwal, A.K.; Khan, S.I.; Heitman, J.; Clark, A.M.; Li, X.C. Synthesis and antifungal activities of miltefosine analogs. Bioorganic Med. Chem. Lett. 2013, 23, 4828–4831. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  80. Widmer, F.; Wright, L.C.; Obando, D.; Handke, R.; Ganendren, R.; Ellis, D.H.; Sorrell, T.C. Hexadecylphosphocholine (miltefosine) has broad-spectrum fungicidal activity and is efficacious in a mouse model of cryptococcosis. Antimicrob. Agents Chemother. 2006, 50, 414–421. [Google Scholar] [CrossRef] [Green Version]
  81. Spadari, C.C.; Vila, T.; Rozental, S.; Ishida, K. Miltefosine has a postantifungal effect and induces apoptosis in Cryptococcus yeasts. Antimicrob. Agents Chemother. 2018, 62, 1–11. [Google Scholar] [CrossRef] [Green Version]
  82. Spadari, C.C.; da Silva de Bastiani, F.W.M.; Lopes, L.B.; Ishida, K. Alginate nanoparticles as non-toxic delivery system for miltefosine in the treatment of candidiasis and cryptococcosis. Int. J. Nanomedicine 2019, 14, 5187–5199. [Google Scholar] [CrossRef] [Green Version]
  83. Wiederhold, N.P.; Najvar, L.K.; Bocanegra, R.; Kirkpatrick, W.R.; Sorrell, T.C.; Patterson, T.F. Limited activity of Miltefosine in murine models of Cryptococcal meningoencephalitis and disseminated Cryptococcosis. Antimicrob. Agents Chemother. 2013, 57, 745–750. [Google Scholar] [CrossRef] [Green Version]
  84. Qian, A.; Zheng, Y.; Wang, R.; Wei, J.; Cui, Y.; Cao, X.; Yang, Y. Design, synthesis, and structure-activity relationship studies of novel tetrazole antifungal agents with potent activity, broad antifungal spectrum and high selectivity. Bioorganic Med. Chem. Lett. 2018, 28, 344–350. [Google Scholar] [CrossRef]
  85. Wang, S.-Q.; Wang, Y.-F.; Xu, Z. Tetrazole hybrids and their antifungal activities. Eur. J. Med. Chem. 2019, 170, 225–234. [Google Scholar] [CrossRef] [PubMed]
  86. Wiederhold, N.P. The antifungal arsenal: Alternative drugs and future targets. Int. J. Antimicrob. Agents 2018, 51, 333–339. [Google Scholar] [CrossRef] [PubMed]
  87. Perfect, J.R. The antifungal pipeline: A reality check. Nat. Rev. Drug Discov. 2017, 16, 603–616. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. Lockhart, S.R.; Fothergill, A.W.; Iqbal, N.; Bolden, C.B.; Grossman, N.T.; Garvey, E.P.; Brand, S.R.; Hoekstra, W.J.; Schotzinger, R.J.; Ottinger, E.; et al. Potent In Vitro activity against Cryptococcus neoformans and Cryptococcus gattii. Antimicrob. Agents Chemother. 2016, 60, 2528–2531. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  89. Nielsen, K.; Vedula, P.; Smith, K.D.; Meya, D.B.; Garvey, E.P.; Hoekstra, W.J.; Schotzinger, R.J.; Boulware, D.R. Activity of VT-1129 against Cryptococcus neoformans clinical isolates with high fluconazole MICs. Med. Mycol. 2017, 55, 453–456. [Google Scholar] [PubMed] [Green Version]
  90. Wiederhold, N.P.; Patterson, H.P.; Tran, B.H.; Yates, C.M.; Schotzinger, R.J.; Garvey, E.P. Fungal-specific Cyp51 inhibitor VT-1598 demonstrates in vitro activity against Candida and Cryptococcus species, endemic fungi, including Coccidioides species, Aspergillus species and Rhizopus arrhizus. J. Antimicrob. Chemother. 2018, 73, 404–408. [Google Scholar] [CrossRef] [Green Version]
  91. Garvey, E.P.; Sharp, A.D.; Warn, P.A.; Yates, C.M.; Schotzinger, R.J. The novel fungal CYP51 inhibitor VT-1598 is efficacious alone and in combination with liposomal amphotericin B in a murine model of cryptococcal meningitis. J. Antimicrob. Chemother. 2018, 73, 2815–2822. [Google Scholar] [CrossRef]
  92. Rossi, S.A.; de Oliveira, H.C.; Agreda-Mellon, D.; Lucio, J.; Mendes Giannini, M.J.S.; García-Cambero, J.P.; Zaragoza, O. Identification of off-patent drugs that show synergism with Amphotericin B or that present antifungal action against Cryptococcus neoformans and Candida spp. Antimicrob. Agents Chemother. 2020, 64, e01921-19. [Google Scholar] [CrossRef]
  93. Joffe, L.S.; Schneider, R.; Lopes, W.; Azevedo, R.; Staats, C.C.; Kmetzsch, L.; Schrank, A.; Del Poeta, M.; Vainstein, M.H.; Rodrigues, M.L. The anti-helminthic compound mebendazole has multiple antifungal effects against Cryptococcus neoformans. Front. Microbiol. 2017, 8, 1–17. [Google Scholar] [CrossRef]
  94. Nixon, G.L.; McEntee, L.; Johnson, A.; Farrington, N.; Whalley, S.; Livermore, J.; Natal, C.; Washbourn, G.; Bibby, J.; Berry, N.; et al. Repurposing and reformulation of the antiparasitic agent flubendazole for treatment of Cryptococcal meningoencephalitis, a neglected fungal disease. Antimicrob. Agents Chemother. 2018, 62, e01909-17. [Google Scholar] [CrossRef] [Green Version]
  95. Nosanchuk, J.D.; Ovalle, R.; Casadevall, A. Glyphosate inhibits melanization of Cryptococcus neoformans and prolongs survival of mice after systemic Infection. J. Infect. Dis. 2001, 183, 1093–1099. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Rodrigues, M.L.; Nakayasu, E.S.; Oliveira, D.L.; Nimrichter, L.; Nosanchuk, J.D.; Almeida, I.C.; Casadevall, A. Extracellular vesicles produced by Cryptococcus neoformans contain protein components associated with virulence. Eukaryot. Cell 2008, 7, 58–67. [Google Scholar] [CrossRef] [Green Version]
  97. Mor, V.; Rella, A.; Farnoud, A.M.; Singh, A.; Munshi, M.; Bryan, A.; Naseem, S.; Konopka, J.B.; Ojima, I.; Bullesbach, E.; et al. Identification of a new class of antifungals targeting the synthesis of fungal sphingolipids. MBio 2015, 6, e00647-15. [Google Scholar] [CrossRef] [Green Version]
  98. Wohlfart, S.; Gelperina, S.; Kreuter, J. Transport of drugs across the blood–brain barrier by nanoparticles. J. Control. Release 2012, 161, 264–273. [Google Scholar] [CrossRef]
  99. Zeeshan, M.; Mukhtar, M.; Ul Ain, Q.; Khan, S.; Ali, H. Nanopharmaceuticals: A boon to the brain-targeted drug delivery. In Pharmaceutical Formulation Design—Recent Practices; IntechOpen: London, UK, 2020; Volume I, pp. 1–21. [Google Scholar]
  100. Zhou, Y.; Peng, Z.; Seven, E.S.; Leblanc, R.M. Crossing the blood–brain barrier with nanoparticles. J. Control. Release 2018, 270, 290–303. [Google Scholar] [CrossRef] [PubMed]
  101. Niu, X.; Chen, J.; Gao, J. Nanocarriers as a powerful vehicle to overcome blood–brain barrier in treating neurodegenerative diseases: Focus on recent advances. Asian J. Pharm. Sci. 2019, 14, 480–496. [Google Scholar] [CrossRef] [PubMed]
  102. Hamidi, M.; Azadi, A.; Rafiei, P. Hydrogel nanoparticles in drug delivery. Adv. Drug Deliv. Rev. 2008, 60, 1638–1649. [Google Scholar] [CrossRef] [PubMed]
  103. Gidwani, M.; Singh, A. V Nanoparticle enabled drug delivery across the blood brain barrier: In vivo and in vitro models, opportunities and challenges. Curr. Pharm. Biotechnol. 2014, 14, 1201–1212. [Google Scholar] [CrossRef]
  104. Garcia-Garcia, E.; Andrieux, K.; Gil, S.; Couvreur, P. Colloidal carriers and blood–brain barrier (BBB) translocation: A way to deliver drugs to the brain? Int. J. Pharm. 2005, 298, 274–292. [Google Scholar] [CrossRef]
  105. Collnot, E.-M.; Baldes, C.; Schaefer, U.F.; Edgar, K.J.; Wempe, M.F.; Lehr, C.-M. Vitamin E TPGS P-Glycoprotein inhibition mechanism: Influence on conformational flexibility, intracellular ATP levels, and role of time and site of access. Mol. Pharm. 2010, 7, 642–651. [Google Scholar] [CrossRef]
  106. Kreuter, J. Nanoparticulate systems for brain delivery of drugs. Adv. Drug Deliv. Rev. 2012, 64, 213–222. [Google Scholar] [CrossRef]
  107. Santangelo, R.; Paderu, P.; Delmas, G.; Chen, Z.-W.; Mannino, R.; Zarif, L.; Perlin, D.S. Efficacy of oral cochleate-Amphotericin B in a mouse model of systemic Candidiasis. Antimicrob. Agents Chemother. 2000, 44, 2356–2360. [Google Scholar] [CrossRef] [Green Version]
  108. Lu, R.; Hollingsworth, C.; Qiu, J.; Wang, A.; Hughes, E.; Xin, X.; Konrath, K.M.; Elsegeiny, W.; Park, Y.-D.; Atakulu, L.; et al. Efficacy of oral encochleated Amphotericin B in a mouse model of Cryptococcal meningoencephalitis. MBio 2019, 10, e00724-19. [Google Scholar] [CrossRef] [Green Version]
  109. Ren, T.; Xu, N.; Cao, C.; Yuan, W.; Yu, X.; Chen, J.; Ren, J. Preparation and therapeutic efficacy of polysorbate-80-coated Amphotericin B/PLA-b-PEG Nanoparticles. J. Biomater. Sci. Polym. 2009, 20, 1369–1380. [Google Scholar] [CrossRef] [Green Version]
  110. Xu, N.; Gu, J.; Zhu, Y.; Wen, H.; Ren, Q.; Chen, J. Efficacy of intravenous amphotericin B-polybutylcyanoacrylate nanoparticles against cryptococcal meningitis in mice. Int. J. Nanomed. 2011, 6, 905–913. [Google Scholar] [CrossRef] [Green Version]
  111. Shao, K.; Huang, R.; Li, J.; Han, L.; Ye, L.; Lou, J.; Jiang, C. Angiopep-2 modified PE-PEG based polymeric micelles for amphotericin B delivery targeted to the brain. J. Control. Release 2010, 147, 118–126. [Google Scholar] [CrossRef] [PubMed]
  112. Shao, K.; Wu, J.; Chen, Z.; Huang, S.; Li, J.; Ye, L.; Lou, J.; Zhu, L.; Jiang, C. A brain-vectored angiopep-2 based polymeric micelles for the treatment of intracranial fungal infection. Biomaterials 2012, 33, 6898–6907. [Google Scholar] [CrossRef]
  113. Ćurić, A.; Keller, B.L.; Reul, R.; Möschwitzer, J.; Fricker, G. Development and lyophilization of itraconazole loaded poly(butyl cyanoacrylate) nanospheres as a drug delivery system. Eur. J. Pharm. Sci. 2015, 78, 121–131. [Google Scholar] [CrossRef] [PubMed]
  114. Ćurić, A.; Möschwitzer, J.P.; Fricker, G. Development and characterization of novel highly-loaded itraconazole poly(butyl cyanoacrylate) polymeric nanoparticles. Eur. J. Pharm. Biopharm. 2017, 114, 175–185. [Google Scholar] [CrossRef] [PubMed]
  115. Zhang, S.; Asghar, S.; Yang, L.; Hu, Z.; Chen, Z.; Shao, F.; Xiao, Y. Borneol and poly (ethylene glycol) dual modified BSA nanoparticles as an itraconazole vehicle for brain targeting. Int. J. Pharm. 2020, 575, 119002. [Google Scholar] [CrossRef]
  116. Zhang, Q.-L.; Fu, B.M.; Zhang, Z.-J. Borneol, a novel agent that improves central nervous system drug delivery by enhancing blood–brain barrier permeability. Drug Deliv. 2017, 24, 1037–1044. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Chen, W.; Zhan, C.; Gu, B.; Meng, Q.; Wang, H.; Lu, W.; Hou, H. Targeted brain delivery of itraconazole via RVG29 anchored nanoparticles. J. Drug Target. 2011, 19, 228–234. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Conventional antifungals and new molecules for cryptococcosis treatment. Amphotericin B (AMB) and azoles inhibit ergosterol and its biosynthesis, respectively, and flucytosine inhibits the nucleic acids synthesis. New molecules acting on non-conventional targets or different structures of fungal cells may have antifungal effects. Erg11 (or Cyp51)—cytochrome P450-dependent lanosterol C14-alpha-demethylase; AcS—Acetyl CoA synthetase; Hsp90—Heat shock protein 90.
Figure 1. Conventional antifungals and new molecules for cryptococcosis treatment. Amphotericin B (AMB) and azoles inhibit ergosterol and its biosynthesis, respectively, and flucytosine inhibits the nucleic acids synthesis. New molecules acting on non-conventional targets or different structures of fungal cells may have antifungal effects. Erg11 (or Cyp51)—cytochrome P450-dependent lanosterol C14-alpha-demethylase; AcS—Acetyl CoA synthetase; Hsp90—Heat shock protein 90.
Microorganisms 08 00613 g001
Figure 2. Schematic representation of nanocarriers, their surface modification and possible routes of transport across the blood–brain barrier (BBB). (A) Nanocarriers frequently employed for drug delivery to the central nervous system. (B) Examples of nanocarrier surface modification to improve passage through the BBB; (C) Possible routes of nanocarrier-mediated transport across the BBB. 1—Nanocarrier-mediated transport; 2—Paracellular pathway, which can result from the ability of a nanocarrier and/or its components to open tight junctions; 3—Adsorption-mediated transcytosis; 4—Transcellular pathway, which might result from the ability of nanocarrier components to improve membrane permeability; 5—Receptor-mediated transcytosis. PEG: polyethyleneglycol; vitE TPGS: D-α-tocopherol polyethylene glycol 1000 succinate.
Figure 2. Schematic representation of nanocarriers, their surface modification and possible routes of transport across the blood–brain barrier (BBB). (A) Nanocarriers frequently employed for drug delivery to the central nervous system. (B) Examples of nanocarrier surface modification to improve passage through the BBB; (C) Possible routes of nanocarrier-mediated transport across the BBB. 1—Nanocarrier-mediated transport; 2—Paracellular pathway, which can result from the ability of a nanocarrier and/or its components to open tight junctions; 3—Adsorption-mediated transcytosis; 4—Transcellular pathway, which might result from the ability of nanocarrier components to improve membrane permeability; 5—Receptor-mediated transcytosis. PEG: polyethyleneglycol; vitE TPGS: D-α-tocopherol polyethylene glycol 1000 succinate.
Microorganisms 08 00613 g002

Share and Cite

MDPI and ACS Style

Spadari, C.d.C.; Wirth, F.; Lopes, L.B.; Ishida, K. New Approaches for Cryptococcosis Treatment. Microorganisms 2020, 8, 613. https://doi.org/10.3390/microorganisms8040613

AMA Style

Spadari CdC, Wirth F, Lopes LB, Ishida K. New Approaches for Cryptococcosis Treatment. Microorganisms. 2020; 8(4):613. https://doi.org/10.3390/microorganisms8040613

Chicago/Turabian Style

Spadari, Cristina de Castro, Fernanda Wirth, Luciana Biagini Lopes, and Kelly Ishida. 2020. "New Approaches for Cryptococcosis Treatment" Microorganisms 8, no. 4: 613. https://doi.org/10.3390/microorganisms8040613

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