3,631
Views
36
CrossRef citations to date
0
Altmetric
Review

Antifungal therapeutics for dimorphic fungal pathogens

&
Pages 211-221 | Received 28 Jul 2016, Accepted 07 Sep 2016, Published online: 01 Nov 2016

ABSTRACT

Dimorphic fungi cause several endemic mycoses which range from subclinical respiratory infections to life-threatening systemic disease. Pathogenic-phase cells of Histoplasma, Blastomyces, Paracoccidioides and Coccidioides escape elimination by the innate immune response with control ultimately requiring activation of cell-mediated immunity. Clinical management of disease relies primarily on antifungal compounds; however, dimorphic fungal pathogens create a number of challenges for antifungal drug therapy. In addition to the drug toxicity issues known for current antifungals, barriers to efficient drug treatment of dimorphic fungal infections include natural resistance to the echinocandins, residence of fungal cells within immune cells, the requirement for systemic delivery of drugs, prolonged treatment times, potential for latent infections, and lack of optimized standardized methodology for in vitro testing of drug susceptibilities. This review will highlight recent advances, current therapeutic options, and new compounds on the horizon for treating infections by dimorphic fungal pathogens.

Introduction

Within the Ascomycetes phylum, members of the genera Histoplasma, Blastomyces, Coccidioides, and Paracoccidioides are dimorphic fungi which are responsible for hundreds of thousands of infections, many thousands of which require clinical intervention. Given similarities in their pathogenesis, the disease manifestations they produce, their clinical management, and particularly the resemblance of their antifungal susceptibility profiles, these 4 genera will be the subject of this review. Two additional dimorphic fungi, Talaromyces marneffei and Sporothrix schenkii, will not be covered. Neither dimorphic fungi nor human pathogens are monophyletic groups within the Onygenales as closely related non-pathogenic and non-dimorphic species exist for several of these genera. While once designated as a single Coccidioides species, Coccidioides immitis, advances in sequence analyses and phylogenetics have separated it into 2 species: Coccidioides immitis and Coccidioides posadasii.Citation1 Other dimorphic fungi have similarly been separated into different species with Paracoccidioides braziliensis now comprised of Paracoccidioides brasiliensis and Paracoccidioides lutziiCitation2 and Blastomyces dermatitidis as the 2 species Blastomyces dermatitidis and Blastomyces gilchristii.Citation3 From a clinical standpoint, management of these diseases does not rely on knowing the precise species. Thus, throughout this review, the dimorphic fungal pathogens will be referred to by their genus name without regard to the specific taxonomic species designation. Both current antifungals and potential new drugs for treating infections by dimorphic fungal pathogens will be discussed.

Dimorphism and endemicity

The hallmark of the dimorphic fungal pathogens is their dual, yet distinct lifestyles. These lifestyles are correlated with separate fungal morphologies with filamentous growth (i.e., hyphae) characterizing the saprobic phase in the environment and yeasts (or spherules in the case of Coccidioides) characterizing the human pathogenic phase. This contrasts with polymorphic fungi (e.g. Candida albicans) which exhibit both yeast and hyphal morphologies in human tissues. Temperature appears to be the central factor that determines the form/lifestyle of the dimorphic fungi with exposure to mammalian body temperature triggering adoption of the pathogenic mode.Citation4-7 Yeasts of the dimorphic fungi (or Coccidoides spherules) are not efficiently eliminated by immune cells. Preventing the transition into yeasts through pharmacologicCitation8,9 or genetic interventionsCitation10-13 renders dimorphic fungal pathogens avirulent indicating the necessity of the dimorphic transition for pathogenesis. Unlike many parasites which must cycle between human and non-human environments to complete their lifecycle, the dimorphic fungi have no such requirement for infection of mammalian hosts. Rather, infection of mammals by the dimorphic fungi is accidental, yet when these fungi transition to the yeast/spherule phase, they express efficient mechanisms that enable their survival and proliferation within this secondary environment.

The dimorphic fungal pathogens are endemic to particular geographic regions rather than being found ubiquitously in the environment. These areas have been largely defined by clinical case prevalence,Citation14 isolation from soils,Citation15 or skin-reactivity tests to specific antigens of the dimorphic fungi.Citation16-18 Ecological aspects of soils that favor growth of the hyphal forms and/or animal or bird patterns for dispersal are thought to underlie dimorphic fungal pathogen endemicity. Histoplasma is endemic to the Midwest and Eastern parts of the United States (chiefly around the Ohio and Mississippi River valleys),Citation16 and is also found in Latin America (particularly Brazil, Venezuela, Colombia, and Argentina;Citation19), parts of Africa,Citation20 and some cases have been reported in ChinaCitation21,22 and India.Citation19 Blastomyces overlaps with many of the Histoplasma-endemic regions in the United States (Midwest, Southeast, and South-central) and also includes Canadian provinces around the Great LakesCitation23,24 and parts of Africa.Citation25 In contrast to the moist environments with decaying organic material supporting Histoplasma and Blastomyces, Coccidioides is found in the more arid environments of the Southwest of the United States (Southern California, Arizona, New Mexico, Texas;Citation26), Central America, and parts of South America (Northern Brazil, Venezuela, Argentina, and ParaguayCitation27). Paracoccidioides species are endemic to South America, particularly areas of Brazil, Colombia, Venezuela, Argentina, and Uruguay.Citation28

The annual incidence of infections by dimorphic fungi is likely inaccurate as under-diagnosis and under-reporting of infections is common. Furthermore, most infections are self limiting without requiring clinical intervention. Nevertheless, estimates of over a half million infections by Histoplasma and Coccidioides occur each year.Citation29,30 Paracoccidioides and Blastomyces infection estimates are more difficult due to regional differences, however reports suggest the incidence ranges from 0.05 to 3 per 100,000 individuals in endemic regions.Citation23,31-33 Together these 4 dimorphic fungal pathogens infect hundreds of thousands each year, a number which does not include veterinary infections. The number of clinical cases typically range from 5–30% of infections, although in some outbreaks over 50% of infections can result in clinical disease.Citation34-36 One survey of hospital records in the United States tallied over 6000 hospitalizations in one year: 3360 cases of histoplasmosis, 771 cases of blastomycosis, and 2194 cases of coccidiodomycosis.Citation14 Less than half of these were in immunocompromised individuals. A case series analysis of Latin America countries showed over 750 annual reported cases of paracoccidioidomycosis.Citation28

Dimorphic fungal pathogen disease and treatment

Inhalation of conidia initiates infection by the dimorphic fungiCitation28,37-39 The conidia (or arthroconidia in the case of Coccidioides) are produced solely by the environmental mycelia. The small size of these propagules facilitates aerosolization upon environmental disturbance and deposition into alveoli once inhaled. Thus, occupational and recreational activities that contribute to soil and environmental disruption are a primary risk factor for infection by dimorphic fungal pathogens.Citation39-42 Most infection outbreaks can be traced back to specific events that led to conidia release from soils and their subsequent inhalation.Citation34,35,43-47 Therefore, practices to avoid or reduce the potential for aerosolization and inhalation of conidia are prescribed for high risk areasCitation34,41,48,49

Within the lungs, conidia transition into pathogenic-phase cells which survive innate immune defenses. For Histoplasma, Blastomyces, and Paracoccidioides, the conidia germinate into yeasts. The yeasts can be internalized by host phagocytes (e.g.,, alveolar macrophages) to varying degrees with Histoplasma yeasts residing almost exclusively within these host cells. This intracellular residence creates an additional barrier to antifungal drug penetration which must be considered in antifungal development. For Coccidioides, the inhaled arthroconidia become spherules that are exclusively extracellular. The spherules enlarge into endosporulating structures within the lung tissue and release endospores for propagation of the infection. Whether phagocytosed or not, the dimorphic fungal pathogens are not controlled by the innate axis of the immune system unlike the opportunistic fungal pathogens. Thus, Histoplasma, Blastomyces, Coccidioides species, and Paracoccidioides infections are not restricted to immunocompromised hosts, but also cause disease in immunocompetent individuals. Control of the infection requires activation of CD4+ cells and consequently individuals lacking aspects of cellular immunity (e.g., HIV, immunosuppression due to tissue or organ transplantation, TNFα blockade, etc.) typically progress to severe and disseminated disease. Elimination of symptoms has been assumed to indicate clearance of the infection, but evidence is now suggesting that at least in some individuals, the infection can enter a latent state, which can re-emerge later when the balance between pathogen and host immunity is altered (e.g., immunosuppression of the host).Citation50-56

Since inhalation is the route of exposure, mycoses caused by dimorphic fungal pathogens are initially pulmonary diseases. In immunocompetent individuals, mild disease is mostly subclinical, often going undiagnosed. Infection causes varying degrees of pneumonia and influenza-like symptoms. For the majority of individuals, symptoms typically resolve without requiring intervention. Roughly 5% of Histoplasma infections are estimated to require clinical managementCitation57 and up to 30% for Coccidioides infections.Citation58 For individuals inhaling a larger inoculum, or those that have some deficiency in cellular immune response, disease is more severe and the infection typically disseminates to extrapulmonary sites via the hemolymphatic system. Extrapulmonary disease presentation varies, but can include oral and pharyngeal mucosa (Paracoccidioides), cutaneous lesions (Histoplasma, Blastomyces, Paracoccidioides, and Coccidioides), and bone (Blastomyces).Citation31,38-40,59 The systemic nature of infections by dimorphic fungi thus precludes any topical antifungal options for management, and the required systemic drug administration significantly elevates the potential for problems with host toxicities.

Current antifungal options

In vitro activities of current antifungal drugs used clinically have been established for the dimorphic fungi. These include drugs of the polyene, azole, and echinocandin classes. In general, reference methods for antimicrobial susceptibility tests follow those established by the Clinical and Laboratory Standards Institute (CLSI). Notably missing from these reference methods are procedures specifically designated for dimorphic fungi leading to confusion as to whether macro- and microdilution test methods for yeasts (M27-A3)Citation60 or for filamentous fungi (M38-A2)Citation61 should be followed. This is not an insignificant question as the yeast and hyphal forms of the dimorphic fungi can have dramatically different susceptibilities.Citation62-69 For example, early studies with echinocandins showed antifungal effects on the hyphal form of Histoplasma.Citation67 However, subsequent tests against the yeast form indicated that caspofungin was anywhere from 20- to 1000-fold less effective.Citation64,68,69 It is now well recognized that the echinocandins have poor antifungal activity on pathogenic phase cells of the dimorphic fungi. This situation underscores the need to use the form most relevant to human infection when establishing the antifungal susceptibility profile of dimorphic fungal pathogens. Unfortunately, some antifungal studies continue to ignore the pathogenic forms and instead limit studies to mycelial-phase cells.Citation70-72 Further complicating antifungal susceptibility profiling are the lack of standardized methods addressing complications for dimorphic fungi. While the CLSI methods for yeasts (M27-A3)Citation60 work well for Candida and Cryptococcus yeasts, they are inadequate for testing yeast-phase cells of the dimorphic fungal pathogens, which have longer generation times and require higher inocula for efficient and consistent growth in broth culture. A more appropriate microdilution method has recently been established for antifungal testing of Histoplasma yeasts,Citation69 and this methodology produces more accurate data for Histoplasma susceptibility.Citation73

Despite these caveats, a general profile of the antimicrobial susceptibilities of dimorphic fungi has been developed for amphotericin B and azole-class drugs. Each of the dimorphic fungal pathogens exhibit a similar susceptibility profile for current antifungals with both amphotericin B and azole drugs generally showing potent activity (low minimum inhibitory concentrations; MIC) with some variability in MICs reported for fluconazole (). Unfortunately, the echinocandins, which have dramatically decreased host toxicity, have poor antifungal activity against the pathogenic-phase cells of dimorphic fungal pathogen yeast cells. The reasons underlying this natural resistance of yeasts to the β-glucan synthase inhibitors is currently unknown.

Table 1. In vitro antifungal MICs for dimorphic fungal pathogens.

The in vitro antifungal susceptibilities for amphotericin B and azole-class drugs have been validated in murine models of dimorphic fungal disease and/or in clinical trials. As a consequence, the Infectious Disease Society of America has released treatment guidelines for infections with Histoplasma,Citation74 Blastomyces,Citation75 and Coccidioides.Citation76 Despite its potential for host toxicity, amphotericin B is recommended for severe disseminated disease, with one of the liposomal formulations preferred. For less severe situations, for follow up after amphotericin B, or for possible prophylaxis of highly susceptible individuals (e.g. AIDS), IDSA guidelines recommend treatment with an oral azole (i.e., itraconazole) with monitoring of serum concentrations to ensure sufficient absorption and bioavailability. To ensure sufficient clearance of the dimorphic fungal infection, treatments typically involve protracted regimens. For mild disease, treatment durations can range from several months to a year depending on the specific dimorphic fungus. Treatment of disseminated disease and disease in immunocompromised hosts can be a year or longer. Beyond the prolonged treatment times, the potential for latency, rather than clearance of the infection, further complicates antifungal management of dimorphic fungal infections.

New and alternative antifungal developments

With the known host toxicity risks of current antifungals,Citation77-79 and the endogenous resistance of dimorphic fungi to the lower toxicity echinocandins, new and alternative antifungal drug options have been explored. The eukaryotic nature shared by both the dimorphic fungal pathogens and the host is recognized as a significant obstacle to antifungal drug development. Consequently, many molecules with promising antifungal activity fail due to lack of adequate selectivity.

One approach to improve the selectivity of existing antifungal molecules, is to test modifications to the structure that decrease drug affinity to host targets. The oral availability of azole-class antifungals is a great benefit to management of dimorphic fungal infections. However, the heme iron-binding action of the imidazoles and triazoles that inhibits fungal cytochrome CYP51 (Erg11) also targets host P450 cytochromes leading to hepatotoxicity and problematic drug-drug interactions. Exploration of other metal binding azole groups in combination with structure-guided improvement of affinity for fungal CYP51 outside of the active site led to the design of compound VT-1161.Citation80 As predicted from structure affinity calculations, the molecule has low affinity for mammalian CYP3A4 but is highly potent against C. albicans. Subsequent testing against Coccidioides arthroconidia in vitro showed VT-1161 has an MIC around 1.5 μg/mL.Citation81 Oral administration of VT-1161 in a murine model of coccidioidomycosis demonstrated VT-1161 reduced spherule burden in the lungs by 100- to 1000-fold, leading to improved mouse survival.Citation81 While VT-1161 had similar effectiveness as fluconazole in vivo, it is hoped the better selectivity of the molecules will decrease potential host toxicity.

Following the goal of reduced host toxicity, some antibacterial compounds have been investigated due to their established safety profile to the host. Drugs targeting the folate pathway have been effective against microbes from bacteria to parasites,Citation82 are orally available, and have a moderately safe host toxicity profile. These drugs have already been a means of prophylaxis against Pneumocystis fungal infections in HIV+ individuals. Sulfonamides in combination with dihydrofolate reductase inhibitors (e.g., cotrimoxazole) have activity against Histoplasma yeasts (8–14 μg/mL and 2–3 μg/mL, respectively)Citation65 and Coccidioides (MICs of 1000 ug/mL and 200 ug/mL, respectively),Citation66 although only the mycelial phase has been tested for Coccidioides. The sulfonamides/antifolate combination has been one of the traditional treatments for ParacoccidioidesCitation59,83-85 but they have not been advanced clinically for the other dimorphic fungal pathogens.

Using a similar rationale, ciprofloxacin, a fluoroquinolone antibacterial with very low host toxicity, has been tested for antifungal potential. The fluoroquinolones inhibit bacterial type II topoisomerases (e.g., DNA gyrase) and were suggested to potentially also inhibit fungal topoisomerase. While not effective against mycelial forms of Histoplasma and Coccidioides, tests against Histoplasma yeast showed ciprofloxacin has an in vitro MIC of 62.5–250 μg/mL.Citation86 While not considered a low MIC, the lack of significant host toxicity for ciprofloxacin creates a good selectivity index and raises its potential, at least as an adjunct therapy for fungal infections. Addition of ciprofloxacin to amphotericin B or itraconazole indicated some additive effects could be gained through drug combinations.

Anti-tuberculosis drugs show some, albeit low potency, antifungal activity for dimorphic fungal pathogens. When tested in vitro against Coccidioides mycelia, rifampicin, isoniazid, and ethambutol had very high MICs (8500 μg/mL, 500 μg/mL, and 2500 μg/mL, respectivelyCitation72). Effectiveness against spherules was not determined but tests against Histoplasma yeasts similarly had high MICsCitation71 arguing these antibacterial compounds lack any significant antifungal utility. However, using isoniazid as the core, chemical structure modification improved its antifungal potential; 3 of 9 isoniazid derivatives had significantly lower MICs against Histoplasma yeasts with one having inhibitory activity at concentrations as low as 7.8 μg/mL.Citation62 Extending the in vitro tests of the isoniazid-hydrazone derivative to Coccidioides demonstrated it had an MIC of 50 μg/mL against mycelia of this dimorphic fungal pathogen.Citation87 Although Coccidioides spherules were not tested, there is a reasonable possibility that the MIC will be lower since Histoplasma yeasts were 8–16X more sensitive to the isoniazid-hydrazone than were mycelia.Citation62

Antiretroviral protease inhibitors have inhibitory activity against Histoplasma. Saquinavir is active against both filamentous and yeast forms with an MIC around 0.4 μg/mL.Citation63 Another protease inhibitor, ritonavir, has an MIC of 1.0 μg/mL against Histoplasma mycelia but is 7-fold more potent against yeast cells. As HIV+ individuals comprise a significant portion of clinical Histoplasma cases, this raises the possibility that management of HIV through protease inhibitor cocktails could simultaneously directly combat infection by dimorphic fungal pathogens.

Surprisingly, farnesol can inhibit growth and morphological transitions of Paracoccidioides and Coccidioides. Farnesol is an alcohol present in some essential oils but is also produced by C. albicans as a quorum sensing molecule. When tested on Paracoccidioides yeasts, farnesol had an MIC of 30 μM.Citation88 Farnesol also inhibited the transition from yeast cells to hyphae, although this morphological defect has questionable relevance to the situation in mammalian hosts. Against Coccidioides mycelia, farnesol was nearly 100-fold more potent (MIC approximately 0.3 μM;Citation70). Farnesol was also reported to inhibit Histoplasma yeast growth with an average MIC of 0.02 μM.Citation89 As a caution, tests with Histoplasma yeasts followed the CLSI methodology (M27-A3) and this has been shown to be inferior for reliable testing of Histoplasma yeast susceptibility.Citation69

Repurposing of anti-cancer drugs has also been fruitful in the search for new antifungal prospects. AR-12 is a celecoxib-derivative that has been shown to have antimicrobial properties against microbes ranging from bacteria to parasites to fungi and even viruses.Citation90-94 Early mechanistic studies in cancer cells suggested AR-12 inhibited cellular phosphoinositide-dependent kinase-1 (Pdk1), however this does not appear to be the case in fungi. Recently, it was shown that AR-12 inhibits acetyl-CoA synthetase, an essential enzyme in fungi.Citation95 AR-12 has antifungal activity against a broad collection of fungi, including the dimorphic fungal pathogens at concentrations around 4–8 μg/mL.Citation73 In vitro, AR-12 is fungicidal against Histoplasma yeasts (Rappleye CA, personal communication). These antifungal characteristics combined with AR-12s safety as established in Phase I trials make AR-12 an attractive antifungal candidate for further development.

Both phenotypic and target-based screening approaches have been used to identify novel compounds that inhibit dimorphic fungal pathogens. Genome analyses for potential antifungal targets focused on those genes that were (i) demonstrated to be essential for fungal growth and (ii) conserved among diverse fungi (Basidiomycetes to Ascomycetes;Citation96). From this thioredoxin reductase (Trr) was selected as a candidate target for computational screening of a library of 3000 compounds. Three compounds were identified and tests against purified Paracoccidioides Trr protein confirmed the compound inhibited Trr enzymatic activity.Citation97 Phenotype tests demonstrated that 2 compounds had significant biological activity against Paracoccidioides yeasts with MICs of 8–16 μg/mL (compounds F1806-0122 and F3307-0100). This study validates target based screening as an approach to identification of potential antifungal compounds.

Phenotypic screening of small molecule libraries has identified a group of aminothiazoles with antifungal activity toward Histoplasma. To enable rapid quantitative screening of yeast growth, yeasts were engineered to express a red-fluorescent protein, which could be used as a surrogate indicator of the number of yeast cells.Citation98 Furthermore, fluorescence-based growth analysis facilitated efficient monitoring of Histoplasma within macrophages, and thus the determination of the effectiveness of compounds on inhibition of Histoplasma within its host cell environment. The top hit aminothiazole (41F5) was effective against yeasts in culture (MIC 2–4 μM) and effectively inhibited proliferation of yeasts resident within macrophages.Citation98 Importantly, the aminothiazole compound had low toxicity to cultured mammalian cells, including macrophages. Unfortunately, the aminothiazole was not active against Blastomyces yeasts despite the very close phylogenetic relationship between Histoplasma and Blastomyces. The molecular target of the aminothiazole remains to be determined, and this may facilitate structure optimization to broaden its activity for other dimorphic fungal pathogens.

The most advanced new antifungal option for clinical development to manage dimorphic fungal pathogens is nikkomycin Z. The polyoxins (from which the nikkomycins are derived) are peptide modified-nuceloside analogs which were originally identified by screening Streptomyces products for antifungal and insecticidal activities.Citation99 These compounds were later shown to be inhibitors of chitin synthesis.Citation100 Since chitin synthase is absent from mammalian cells, these compounds have very high selectivity for fungi. Following polyoxin studies on C. albicans, the nikkomycins were demonstrated to have good potency in vitro against dimorphic fungal pathogens,Citation101-103 with the most potency against Coccidioides cells (MIC of 0.125 μg/mL for spherules).Citation103 Examination of treated Coccidioides spherules showed lack of endosporulation and even lysis of the spherules consistent with impairments in cell wall structure.Citation103 The nikkomycins are orally available, simplifying extension of studies to animal models of mycoses caused by dimorphic fungal pathogens. Administration of nikkomycin Z in murine models of lethal histoplasmosis resulted in decreased organ fungal burdens (3- to 28-fold) and a concomitant enhancement of mouse survival at doses from 20 to 100 mg/kg per day.Citation102-104 Significant protection from lethal infection was realized, however, only at moderate inocula suggesting nikkomycin antifungal activity may not be sufficiently rapid to curb severe infections. Similar results were found for treatment of blastomycosis with reduction in fungal burdens ranging from 10-fold (at a dose of 50 mg/kg)Citation103 to apparent lung tissue sterilization (over 3500-fold reduction at higher doses of 400–1000 mg/kg).Citation105 The ability to safely administer such doses is a testament to the high selectivity gained by targeting the fungi-specific molecule chitin synthase. Nikkomycin Z is also effective in treating pulmonary coccidioidomycosis with over 10,000-fold reduction in lung fungal burdens and complete protection from lethal infection of mice at a dose of 20 mg/kg.Citation103 Recently, a small clinical trial in dogs with Coccidioides infection was completed. Dogs with mild to moderate disease had significant improvement but those with severe disease had little or poor responses to treatment, again suggesting nikkomycin may not act rapidly enough to quickly curtail disease progression in high fungal burden situations. Nonetheless, nikkomycin Z shows good promise as a highly selective (low host toxicity) antifungal for development against dimorphic fungal pathogen infections.

Conclusions

Current clinical management of infections by dimorphic fungal pathogens is limited to azole-class antifungal drugs and amphotericin B. While orally available, the azoles are not without host toxicity issues and the treatment course is lengthy for infections by dimorphic fungi. Development of resistance to azoles is not widespread, although treatment failures due to azole resistance have occurred.Citation106-108 Unfortunately, the better tolerated echinocandin antifungals lack efficacy against the pathogenic-phase of the dimorphic fungal pathogens raising the need for alternative or second-line treatment options.

While a number of strides have been made in repurposing existing drugs and development of new inhibitors of fungal growth, careful attention must be paid to challenges posed by dimorphic fungi. As the yeasts/spherules of the dimorphic fungi are the state present within the mammalian host, antimicrobial susceptibilities need to be performed with these pathogenic-phase cells, not the mycelia which has led to erroneous conclusions. Testing of the pathogenic-phase in vitro should follow recently optimized procedures as the CLSI methodology for yeasts is inadequate for the dimorphic fungi. Since yeast cells of the dimorphic fungi reside within host phagocytes, it is also advisable for in vitro tests to be followed with tests on drug effectiveness on intracellular yeasts, at least during initial drug development stages.

The overall selectivity of antifungal drug candidates is critical for progression of drugs through the development pipeline. Structure-guided rational design is one approach that has improved the selectivity of an azole structure (VT-1161). Many of the repurposed drugs have relatively high MICs (greater than 100 ug/mL) questioning their therapeutic utility, however if their selectivity is sufficiently high, formulations may be developed to facilitate sufficiently high serum and tissue levels. Lower MICs have been found for drugs targeting the folate pathway, an isoniazid-hydrazone derivative, antiretroviral protease inhibitors, and the anti-cancer drug AR-12, all of which are expected to be reasonably well-tolerated by the mammalian host. Novel drugs with good in vitro MICs and good selectivity include thioredoxin-reductase inhibitors, an aminothiazole compound, and nikkomycin Z. Since nikkomycin Z targets an enzyme absent from the host, nikkomycin has an excellent basis for high selectivity for fungi. In addition, nikkomycin Z has maintained antifungal effectiveness against multiple dimorphic fungal pathogens in animal models of disease. While the current antifungal armament is limited, there are exciting prospects on the horizon for treating dimorphic fungal infections.

Disclosure of potential conflicts of interest

No potential conflicts of interest were disclosed.

Funding

Studies on susceptibility to established drugs and development of new antifungals was supported, in part, by grant R21-AI109437 from the National Institute of Allergy and Infectious Diseases (NIAID).

References

  • Fisher MC, Koenig GL, White TJ, Taylor JW. Molecular and phenotypic description of Coccidioides posadasii sp. nov., previously recognized as the non-California population of Coccidioides immitis. Mycologia 2002; 94:73-84; PMID:21156479; http://dx.doi.org/10.2307/3761847
  • Teixeira MM, Theodoro RC, de Carvalho MJA, Fernandes L, Paes HC, Hahn RC, Mendoza L, Bagagli E, San-Blas G, Felipe MSS. Phylogenetic analysis reveals a high level of speciation in the Paracoccidioides genus. Mol Phylogenet Evol 2009; 52:273-83; PMID:19376249; http://dx.doi.org/10.1016/j.ympev.2009.04.005
  • Brown EM, McTaggart LR, Zhang SX, Low DE, Stevens DA, Richardson SE. Phylogenetic analysis reveals a cryptic species Blastomyces gilchristii, sp. nov. within the human pathogenic fungus Blastomyces dermatitidis. PLoS One 2013; 8:e59237; PMID:23533607; http://dx.doi.org/10.1371/journal.pone.0059237
  • Horwath MC, Fecher RA, Deepe GS. Histoplasma capsulatum, lung infection and immunity. Future Microbiol 2015; 10:967-75; PMID:26059620; http://dx.doi.org/10.2217/fmb.15.25
  • Klein BS, Newman SL. Role of cell-surface molecules of Blastomyces dermatitidis in host-pathogen interactions. Trends Microbiol 1996; 4:246-51; PMID:8795162; http://dx.doi.org/10.1016/0966-842X(96)10028-7
  • Kellner EM, Orsborn KI, Siegel EM, Mandel MA, Orbach MJ, Galgiani JN. Coccidioides posadasii contains a single 1,3-β-glucan synthase gene that appears to be essential for growth. Eukaryotic Cell 2005; 4:111-20; PMID:15643067; http://dx.doi.org/10.1128/EC.4.1.111-120.2005
  • Abreu e Silva MÀ de, Salum FG, Figueiredo MA, Cherubini K. Important aspects of oral paracoccidioidomycosis–a literature review. Mycoses 2013; 56:189-99; PMID:23088400; http://dx.doi.org/10.1111/myc.12017
  • Medoff G, Kobayashi GS, Painter A, Travis S. Morphogenesis and pathogenicity of Histoplasma capsulatum. Infect Immun 1987; 55:1355-8; PMID:3032799
  • Medoff G, Sacco M, Maresca B, Schlessinger D, Painter A, Kobayashi GS, Carratu L. Irreversible block of the mycelial-to-yeast phase transition of Histoplasma capsulatum. Science 1986; 231:476-9; PMID:3001938; http://dx.doi.org/10.1126/science.3001938
  • Nemecek JC, Wüthrich M, Klein BS. Global control of dimorphism and virulence in fungi. Science 2006; 312:583-8; PMID:16645097; http://dx.doi.org/10.1126/science.1124105
  • Nguyen VQ, Sil A. Temperature-induced switch to the pathogenic yeast form of Histoplasma capsulatum requires Ryp1, a conserved transcriptional regulator. Proc Natl Acad Sci U S A 2008; 105:4880-5; PMID:18339808; http://dx.doi.org/10.1073/pnas.0710448105
  • Beyhan S, Gutierrez M, Voorhies M, Sil A. A temperature-responsive network links cell shape and virulence traits in a primary fungal pathogen. PLoS Biol 2013; 11:e1001614; PMID:23935449; http://dx.doi.org/10.1371/journal.pbio.1001614
  • Boyce KJ, Schreider L, Kirszenblat L, Andrianopoulos A. The two-component histidine kinases DrkA and SlnA are required for in vivo growth in the human pathogen Penicillium marneffei. Mol Microbiol 2011; 82:1164-84; PMID:22059885; http://dx.doi.org/10.1111/j.1365-2958.2011.07878.x
  • Chu JH, Feudtner C, Heydon K, Walsh TJ, Zaoutis TE. Hospitalizations for endemic mycoses: a population-based national study. Clin Infect Dis 2006; 42:822-5; PMID:16477560; http://dx.doi.org/10.1086/500405
  • Ajello L. Coccidioidomycosis and histoplasmosis. A review of their epidemiology and geographical distribution. Mycopathol Mycol Appl 1971; 45:221-30; PMID:4950673; http://dx.doi.org/10.1007/BF02051969
  • Edwards LB, Acquaviva FA, Livesay VT, Cross FW, Palmer CE. An atlas of sensitivity to tuberculin, PPD-B, and histoplasmin in the United States. Am Rev Respir Dis 1969; 99(1–132); PMID:5767603
  • Mochi A, Edwards PQ. Geographical distribution of histoplasmosis and histoplasmin sensitivity. Bull World Health Organ 1952; 5:259-91; PMID:14935779
  • Kauffman CA. Histoplasmosis: a clinical and laboratory update. Clin Microbiol Rev 2007; 20:115-32; PMID:17223625; http://dx.doi.org/10.1128/CMR.00027-06
  • Bahr NC, Antinori S, Wheat LJ, Sarosi GA. Histoplasmosis infections worldwide: thinking outside of the Ohio River valley. Curr Trop Med Rep 2015; 2:70-80; PMID:26279969; http://dx.doi.org/10.1007/s40475-015-0044-0
  • Gugnani HC. Histoplasmosis in Africa: a review. Indian J Chest Dis Allied Sci 2000; 42:271-7; PMID:15597674
  • Zhao B, Xia X, Yin J, Zhang X, Wu E, Shi Y, Tong M, Sun S, Gui X, Zhao Z, et al. Epidemiological investigation of Histoplasma capsulatum infection in China. Chin Med J 2001; 114:743-6; PMID:11780341
  • Pan B, Chen M, Pan W, Liao W. Histoplasmosis: a new endemic fungal infection in China? Review and analysis of cases. Mycoses 2013; 56:212-21; PMID:23216676; http://dx.doi.org/10.1111/myc.12029
  • Smith JA, Gauthier G. New Developments in Blastomycosis. Semin Respir Crit Care Med 2015; 36:715-28; PMID:26398538; http://dx.doi.org/10.1055/s-0035-1562898
  • Klein BS, Vergeront JM, Davis JP. Epidemiologic aspects of blastomycosis, the enigmatic systemic mycosis. Semin Respir Infect 1986; 1:29-39; PMID:3317597
  • Carman WF, Frean JA, Crewe-Brown HH, Culligan GA, Young CN. Blastomycosis in Africa. Mycopathologia 1989; 107:25-32; PMID:2682252; http://dx.doi.org/10.1007/BF00437587
  • Edwards PQ, Palmer CE. Prevalence of sensitivity to coccidioidin, with special reference to specific and nonspecific reactions to coccidioidin and to histoplasmin. Dis Chest 1957; 31:35-60; PMID:13384171; http://dx.doi.org/10.1378/chest.31.1.35
  • Laniado-Laborin R. Expanding understanding of epidemiology of coccidioidomycosis in the Western hemisphere. Ann N Y Acad Sci 2007; 1111:19-34; PMID:17395731; http://dx.doi.org/10.1196/annals.1406.004
  • Martinez R. Epidemiology of paracoccidioidomycosis. Rev Inst Med Trop Sao Paulo 2015; 57(19):11-20; PMID:26465364; http://dx.doi.org/10.1590/S0036-46652015000700004
  • Hammerman KJ, Powell KE, Tosh FE. The incidence of hospitalized cases of systemic mycotic infections. Med Mycol 1974; 12:33-45; http://dx.doi.org/10.1080/00362177485380061
  • Stevens DA. Coccidioidomycosis. N Engl J Med 1995; 332:1077-82; PMID:7898527; http://dx.doi.org/10.1056/NEJM199504203321607
  • Saccente M, Woods GL. Clinical and laboratory update on blastomycosis. Clin Microbiol Rev 2010; 23:367-81; PMID:20375357; http://dx.doi.org/10.1128/CMR.00056-09
  • Restrepo A, McEwen JG, Castañeda E. The habitat of Paracoccidioides brasiliensis: how far from solving the riddle? Med Mycol 2001; 39:233-41; PMID:11446526; http://dx.doi.org/10.1080/714031028
  • Colombo AL, Tobón A, Restrepo A, Queiroz-Telles F, Nucci M. Epidemiology of endemic systemic fungal infections in Latin America. Med Mycol 2011; 49:785-98; PMID:21539506
  • Cummings KC, McDowell A, Wheeler C, McNary J, Das R, Vugia DJ, Mohle-Boetani JC. Point-source outbreak of coccidioidomycosis in construction workers. Epidemiol Infect 2010; 138:507-11; PMID:19845993; http://dx.doi.org/10.1017/S0950268809990999
  • Chamany S, Mirza SA, Fleming JW, Howell JF, Lenhart SW, Mortimer VD, Phelan MA, Lindsley MD, Iqbal NJ, Wheat LJ, et al. A large histoplasmosis outbreak among high school students in Indiana, 2001. Pediatr Infect Dis J 2004; 23:909-14; PMID:15602189; http://dx.doi.org/10.1097/01.inf.0000141738.60845.da
  • Klein BS, Vergeront JM, Weeks RJ, Kumar UN, Mathai G, Varkey B, Kaufman L, Bradsher RW, Stoebig JF, Davis JP. Isolation of Blastomyces dermatitidis in soil associated with a large outbreak of blastomycosis in Wisconsin. N Engl J Med 1986; 314:529-34; PMID:3945290; http://dx.doi.org/10.1056/NEJM198602273140901
  • Kauffman CA. Histoplasmosis. Clin Chest Med 2009; 30:217-25; PMID:19375629; http://dx.doi.org/10.1016/j.ccm.2009.02.002
  • Stockamp NW, Thompson GR. Coccidioidomycosis. Infect Dis Clin North Am 2016; 30:229-46; PMID:26739609; http://dx.doi.org/10.1016/j.idc.2015.10.008
  • Castillo CG, Kauffman CA, Miceli MH. Blastomycosis. Infect Dis Clin North Am 2016; 30:247-64; PMID:26739607; http://dx.doi.org/10.1016/j.idc.2015.10.002
  • Wheat LJ, Azar MM, Bahr NC, Spec A, Relich RF, Hage C. Histoplasmosis. Infect Dis Clin North Am 2016; 30:207-27; PMID:26897068; http://dx.doi.org/10.1016/j.idc.2015.10.009
  • Johnson L, Gaab EM, Sanchez J, Bui PQ, Nobile CJ, Hoyer KK, Peterson MW, Ojcius DM. Valley fever: danger lurking in a dust cloud. Microbes Infect 2014; 16:591-600; PMID:25038397; http://dx.doi.org/10.1016/j.micinf.2014.06.011
  • Twarog M, Thompson GR. Coccidioidomycosis: Recent updates. Semin Respir Crit Care Med 2015; 36:746-55; PMID:26398540; http://dx.doi.org/10.1055/s-0035-1562900
  • Carlos WG, Rose AS, Wheat LJ, Norris S, Sarosi GA, Knox KS, Hage CA. Blastomycosis in indiana: digging up more cases. Chest 2010; 138:1377-82; PMID:20558552; http://dx.doi.org/10.1378/chest.10-0627
  • Benedict K, Mody RK. Epidemiology of Histoplasmosis Outbreaks, United States, 1938-2013. Emerging Infect Dis 2016; 22:370-8; PMID:26890817; http://dx.doi.org/10.3201/eid2203.151117
  • Schneider E, Hajjeh RA, Spiegel RA, Jibson RW, Harp EL, Marshall GA, Gunn RA, McNeil MM, Pinner RW, Baron RC, et al. A coccidioidomycosis outbreak following the Northridge, Calif, earthquake. JAMA 1997; 277:904-8; PMID:9062329; http://dx.doi.org/10.1001/jama.1997.03540350054033
  • Crum N, Lamb C, Utz G, Amundson D, Wallace M. Coccidioidomycosis outbreak among United States Navy SEALs training in a Coccidioides immitis-endemic area–Coalinga, California. J Infect Dis 2002; 186:865-8; PMID:12198626; http://dx.doi.org/10.1086/342409
  • Wheat LJ, Slama TG, Eitzen HE, Kohler RB, French ML, Biesecker JL. A large urban outbreak of histoplasmosis: clinical features. Ann Intern Med 1981; 94:331-7; PMID:7224378; http://dx.doi.org/10.7326/0003-4819-94-3-331
  • Das R, McNary J, Fitzsimmons K, Dobraca D, Cummings K, Mohle-Boetani J, Wheeler C, McDowell A, Iossifova Y, Bailey R, et al. Occupational coccidioidomycosis in California: outbreak investigation, respirator recommendations, and surveillance findings. J Occup Environ Med 2012; 54:564-71; PMID:22504958; http://dx.doi.org/10.1097/JOM.0b013e3182480556
  • Histoplasmosis - Protecting Workers at Risk. DHHS (NIOSH) Publication 2004; No. 2005-109. Available from: https://www.cdc.gov/niosh/docs/2005-109/
  • Jain VV, Evans T, Peterson MW. Reactivation histoplasmosis after treatment with anti-tumor necrosis factor α in a patient from a nonendemic area. Respir Med 2006; 100:1291-3; PMID:16545951; http://dx.doi.org/10.1016/j.rmed.2005.09.020
  • Vergidis P, Avery RK, Wheat LJ, Dotson JL, Assi MA, Antoun SA, Hamoud KA, Burdette SD, Freifeld AG, McKinsey DS, et al. Histoplasmosis complicating tumor necrosis factor-α blocker therapy: a retrospective analysis of 98 cases. Clin Infect Dis 2015; 61:409-17; PMID:25870331; http://dx.doi.org/10.1093/cid/civ299
  • Arbeloa-Gutierrez L, Kuberski T, Johnson SM, Sagastibelza I, Alaez JI, Pappagianis D. Reactivation of coccidioidomycosis: a prosthetic joint infection in Spain. Eur J Clin Microbiol Infect Dis 2016; 35:183-6; PMID:26638217; http://dx.doi.org/10.1007/s10096-015-2526-3
  • Nakelchik M, Mangino JE. Reactivation of histoplasmosis after treatment with infliximab. Am J Med 2002; 112:78; PMID:11812415; http://dx.doi.org/10.1016/S0002-9343(01)00945-7
  • Van Koeveringe MP, Brouwer RE. Histoplasma capsulatum reactivation with haemophagocytic syndrome in a patient with chronic lymphocytic leukaemia. Neth J Med 2010; 68:418-21; PMID:21209468
  • Ankobiah WA, Vaidya K, Powell S, Carrasco M, Allam A, Chechani V, Kamholz SL. Disseminated histoplasmosis in AIDS. Clinicopathologic features in seven patients from a non-endemic area. N Y State J Med 1990; 90:234-8; PMID:2348942
  • Keckich DW, Blair JE, Vikram HR, Seville MT, Kusne S. Reactivation of coccidioidomycosis despite antifungal prophylaxis in solid organ transplant recipients. Transplantation 2011; 92:88-93; PMID:21516063; http://dx.doi.org/10.1097/TP.0b013e31821c1df6
  • Hage CA, Azar MM, Bahr N, Loyd J, Wheat LJ. Histoplasmosis: up-to-date evidence-based approach to diagnosis and management. Semin Respir Crit Care Med 2015; 36:729-45; PMID:26398539; http://dx.doi.org/10.1055/s-0035-1562899
  • Thompson GR. Pulmonary coccidioidomycosis. Semin Respir Crit Care Med 2011; 32:754-63; PMID:22167403; http://dx.doi.org/10.1055/s-0031-1295723
  • Marques SA. Paracoccidioidomycosis. Clin Dermatol 2012; 30:610-5; PMID:23068148; http://dx.doi.org/10.1016/j.clindermatol.2012.01.006
  • Clinical and Laboratory Standards Institute. Reference method for broth dilution antifungal susceptibility testing of yeasts; approved standard—3rd ed. CLSI document M27-A3. 3rd ed. Wayne PA: Clinical and Laboratory Standards Institute 2008.
  • Clinical and Laboratory Standards Institute. Reference method for broth dilution antifungal susceptibility testing of filamentous fungi; approved standard—2nd ed. CLSI document M38-A2. 2nd ed. Wayne, PA: Clinical and Laboratory Standards Institute 2008.
  • De Aguiar Cordeiro R, de Farias Marques FJ, de Aguiar Cordeiro R, da Silva MR, Donato Maia Malaquias A, Silva de Melo CV, Mafezoli J, Ferreira de Oliveira M da C, Nogueira Brilhante RS, Gadelha Rocha MF, et al. Synthesis and antifungal activity in vitro of isoniazid derivatives against histoplasma capsulatum var. capsulatum. Antimicrob Agents Chemother 2014; 58:2504-11; PMID:24514090; http://dx.doi.org/10.1128/AAC.01654-13
  • Brilhante RSN, Caetano ÉP, Riello GB, Guedes GM de M, Castelo-Branco D de SCM, Fechine MAB, Oliveira JS de, Camargo ZP de, Mesquita JRL de, Monteiro AJ, et al. Antiretroviral drugs saquinavir and ritonavir reduce inhibitory concentration values of itraconazole against Histoplasma capsulatum strains in vitro. Braz J Infect Dis 2016; 20:155-9; PMID:26748233; http://dx.doi.org/10.1016/j.bjid.2015.11.003
  • Hage CA, Connolly P, Horan D, Durkin M, Smedema M, Zarnowski R, Smith P, Wheat LJ. Investigation of the efficacy of micafungin in the treatment of histoplasmosis using two North American strains of Histoplasma capsulatum. Antimicrob Agents Chemother 2011; 55:4447-50; PMID:21670186; http://dx.doi.org/10.1128/AAC.01681-10
  • Brilhante RSN, Fechine MAB, Cordeiro R de A, Rocha MFG, Ribeiro JF, Monteiro AJ, de Lima RAC, Mesquita JRL, de Camargo ZP, Sidrim JJC. In vitro effect of sulfamethoxazole-trimethoprim against Histoplasma capsulatum var. capsulatum. Antimicrob Agents Chemother 2010; 54:3978-9; PMID:20566767; http://dx.doi.org/10.1128/AAC.00793-10
  • Cordeiro R de A, Astete-Medrano DJ, Marques FJ de F, Andrade HTL, Perdigão Neto LV, Tavares JL, de Lima RAC, Patoilo KKNR, Monteiro AJ, Brilhante RSN, et al. Cotrimoxazole enhances the in vitro susceptibility of Coccidioides posadasii to antifungals. Mem Inst Oswaldo Cruz 2011; 106:1045-8; PMID:22241131; http://dx.doi.org/10.1590/S0074-02762011000800024
  • Espinel-Ingroff A. Comparison of In vitro activities of the new triazole SCH56592 and the echinocandins MK-0991 (L-743,872) and LY303366 against opportunistic filamentous and dimorphic fungi and yeasts. J Clin Microbiol 1998; 36:2950-6; PMID:9738049
  • Nakai T, Uno J, Ikeda F, Tawara S, Nishimura K, Miyaji M. In vitro antifungal activity of Micafungin (FK463) against dimorphic fungi: comparison of yeast-like and mycelial forms. Antimicrob Agents Chemother 2003; 47:1376-81; PMID:12654673; http://dx.doi.org/10.1128/AAC.47.4.1376-1381.2003
  • Goughenour KD, Balada-Llasat JM, Rappleye CA. Quantitative microplate-based growth assay for determination of antifungal susceptibility of histoplasma capsulatum yeasts. J Clin Microbiol 2015; 53:3286-95; PMID:26246483; http://dx.doi.org/10.1128/JCM.00795-15
  • Brilhante RSN, de Lima RAC, Caetano EP, Leite JJG, Castelo-Branco D de SCM, Ribeiro JF, Bandeira T de JPG, Cordeiro R de A, Monteiro AJ, Sidrim JJC, et al. Effect of farnesol on growth, ergosterol biosynthesis, and cell permeability in Coccidioides posadasii. Antimicrob Agents Chemother 2013; 57:2167-70; PMID:23459491; http://dx.doi.org/10.1128/AAC.02457-12
  • Cordeiro R de A, Marques FJ de F, Brilhante RSN, Rocha de Castro E Silva K, Mourão CI, Caetano EP, Fechine MAB, Ribeiro JF, Castelo-Branco D de SCM, de Lima RAC, et al. Synergistic effect of antituberculosis drugs and azoles in vitro against Histoplasma capsulatum var. capsulatum. Antimicrob Agents Chemother 2011; 55:4482-4; PMID:21690286; http://dx.doi.org/10.1128/AAC.01471-10
  • De Aguiar Cordeiro R, Brilhante RSN, Rocha MFG, Fechine MAB, Camargo ZP de, Sidrim JJC. In vitro inhibitory effect of antituberculosis drugs on clinical and environmental strains of Coccidioides posadasii. J Antimicrob Chemother 2006; 58:575-9; PMID:16867997; http://dx.doi.org/10.1093/jac/dkl290
  • Koselny K, Green J, DiDone L, Halterman JP, Fothergill AW, Wiederhold NP, Patterson TF, Cushion MT, Rappleye CA, Wellington M, et al. The celecoxib derivative AR-12 has broad spectrum antifungal activity in vitro and improves the 1 activity of fluconazole in a murine model of cryptococcosis. Antimicrob Agents Chemother 2016; PMID:27645246; http://dx.doi.org/10.1128/AAC.01061-16
  • Wheat LJ, Freifeld AG, Kleiman MB, Baddley JW, McKinsey DS, Loyd JE, Kauffman CA. Clinical Practice Guidelines for the Management of Patients with Histoplasmosis: 2007 Update by the infectious diseases society of America. Clin Infect Dis 2007; 45:807-25; PMID:17806045; http://dx.doi.org/10.1086/521259
  • Chapman SW, Dismukes WE, Proia LA, Bradsher RW, Pappas PG, Threlkeld MG, Kauffman CA, Infectious Diseases Society of America. Clinical practice guidelines for the management of blastomycosis: 2008 update by the infectious diseases society of America. Clin Infect Dis 2008; 46:1801-12; PMID:18462107; http://dx.doi.org/10.1086/588300
  • Galgiani JN, Ampel NM, Blair JE, Catanzaro A, Johnson RH, Stevens DA, Williams PL, Infectious Diseases Society of America. Coccidioidomycosis. Clin Infect Dis 2005; 41:1217-23; PMID:16206093; http://dx.doi.org/10.1086/496991
  • Nett JE, Andes DR. Antifungal Agents: Spectrum of activity, pharmacology, and clinical indications. Infect Dis Clin North Am 2016; 30:51-83; PMID:26739608; http://dx.doi.org/10.1016/j.idc.2015.10.012
  • Cheng JT, Witty RT, Robinson RR, Yarger WE. Amphotericin B nephrotoxicity: increased renal resistance and tubule permeability. Kidney Int 1982; 22:626-33; PMID:7162036; http://dx.doi.org/10.1038/ki.1982.221
  • Bates DW, Su L, Yu DT, Chertow GM, Seger DL, Gomes DR, Dasbach EJ, Platt R. Mortality and costs of acute renal failure associated with amphotericin B therapy. Clin Infect Dis 2001; 32:686-93; PMID:11229835; http://dx.doi.org/10.1086/319211
  • Hoekstra WJ, Garvey EP, Moore WR, Rafferty SW, Yates CM, Schotzinger RJ. Design and optimization of highly-selective fungal CYP51 inhibitors. Bioorg Med Chem Lett 2014; 24:3455-8; PMID:24948565; http://dx.doi.org/10.1016/j.bmcl.2014.05.068
  • Shubitz LF, Trinh HT, Galgiani JN, Lewis ML, Fothergill AW, Wiederhold NP, Barker BM, Lewis ERG, Doyle AL, Hoekstra WJ, et al. Evaluation of VT-1161 for Treatment of Coccidioidomycosis in Murine Infection Models. Antimicrob Agents Chemother 2015; 59:7249-54; PMID:26369964; http://dx.doi.org/10.1128/AAC.00593-15
  • Capasso C, Supuran CT. Sulfa and trimethoprim-like drugs - antimetabolites acting as carbonic anhydrase, dihydropteroate synthase and dihydrofolate reductase inhibitors. J Enzyme Inhib Med Chem 2014; 29:379-87; PMID:23627736; http://dx.doi.org/10.3109/14756366.2013.787422
  • Borges SRC, Silva GMS da, Chambela M da C, Oliveira R de VC de, Costa RLB, Wanke B, Valle ACF do. Itraconazole vs. trimethoprim-sulfamethoxazole: A comparative cohort study of 200 patients with paracoccidioidomycosis. Med Mycol 2014; 52:303-10; PMID:24577007; http://dx.doi.org/10.1093/mmy/myt012
  • Cavalcante R de S, Sylvestre TF, Levorato AD, de Carvalho LR, Mendes RP. Comparison between itraconazole and cotrimoxazole in the treatment of paracoccidiodomycosis. PLoS Negl Trop Dis 2014; 8:e2793; http://dx.doi.org/10.1371/journal.pntd.0002793
  • Restrepo A. Treatment of tropical mycoses. J Am Acad Dermatol 1994; 31:S91-102; PMID:8077517; http://dx.doi.org/10.1016/S0190-9622(08)81277-7
  • Brilhante RSN, Caetano EP, Sidrim JJC, Cordeiro RA, Camargo ZP, Fechine MAB, Lima RAC, Castelo Branco DSCM, Marques FJF, Mesquita JRL, et al. Ciprofloxacin shows synergism with classical antifungals against Histoplasma capsulatum var. capsulatum and Coccidioides posadasii. Mycoses 2013; 56:397-401; PMID:23205615; http://dx.doi.org/10.1111/myc.12025
  • Cordeiro R de A, de Melo CVS, Marques FJ de F, Serpa R, Evangelista AJ de J, Caetano EP, Mafezoli J, de Oliveira M da CF, da Silva MR, Bandeira T de JPG, et al. Synthesis and in vitro antifungal activity of isoniazid-derived hydrazones against Coccidioides posadasii. Microb Pathog 2016; 98:1-5; PMID:27334293; http://dx.doi.org/10.1016/j.micpath.2016.06.022
  • Derengowski LS, De-Souza-Silva C, Braz SV, Mello-De-Sousa TM, Báo SN, Kyaw CM, Silva-Pereira I. Antimicrobial effect of farnesol, a Candida albicans quorum sensing molecule, on Paracoccidioides brasiliensis growth and morphogenesis. Ann Clin Microbiol Antimicrob 2009; 8:13; PMID:19402910; http://dx.doi.org/10.1186/1476-0711-8-13
  • Brilhante RSN, de Lima RAC, Marques FJ de F, Silva NF, Caetano ÉP, Castelo-Branco D de SCM, Bandeira T de JPG, Moreira JLB, Cordeiro R de A, Monteiro AJ, et al. Histoplasma capsulatum in planktonic and biofilm forms: in vitro susceptibility to amphotericin B, itraconazole and farnesol. J Med Microbiol 2015; 64:394-9; PMID:25657300; http://dx.doi.org/10.1099/jmm.0.000030
  • Baxter BK, DiDone L, Ogu D, Schor S, Krysan DJ. Identification, in vitro activity and mode of action of phosphoinositide-dependent-1 kinase inhibitors as antifungal molecules. ACS Chem Biol 2011; 6:502-10; PMID:21294551; http://dx.doi.org/10.1021/cb100399x
  • Hoang KV, Curry H, Collier MA, Borteh H, Bachelder EM, Schlesinger LS, Gunn JS, Ainslie KM. Needle-free delivery of acetalated Dextran-Encapsulated AR-12 protects mice from Francisella tularensis Lethal Challenge. Antimicrob Agents Chemother 2016; 60:2052-62; PMID:26787696; http://dx.doi.org/10.1128/AAC.02228-15
  • Collier MA, Peine KJ, Gautam S, Oghumu S, Varikuti S, Borteh H, Papenfuss TL, Sataoskar AR, Bachelder EM, Ainslie KM. Host-mediated Leishmania donovani treatment using AR-12 encapsulated in acetalated dextran microparticles. Int J Pharm 2016; 499:186-94; PMID:26768723; http://dx.doi.org/10.1016/j.ijpharm.2016.01.004
  • Chiu H-C, Kulp SK, Soni S, Wang D, Gunn JS, Schlesinger LS, Chen C-S. Eradication of intracellular Salmonella enterica serovar Typhimurium with a small-molecule, host cell-directed agent. Antimicrob Agents Chemother 2009; 53:5236-44; PMID:19805568; http://dx.doi.org/10.1128/AAC.00555-09
  • Mohr EL, McMullan LK, Lo MK, Spengler JR, Bergeron É, Albariño CG, Shrivastava-Ranjan P, Chiang CF, Nichol ST, Spiropoulou CF, et al. Inhibitors of cellular kinases with broad-spectrum antiviral activity for hemorrhagic fever viruses. Antiviral Res 2015; 120:40-7; PMID:25986249; http://dx.doi.org/10.1016/j.antiviral.2015.05.003
  • Koselny K, Green J, Favazzo L, Glazier VE, DiDone L, Ransford S, Krysan DJ. Antitumor/Antifungal Celecoxib Derivative AR-12 is a non-nucleoside inhibitor of the ANL-Family Adenylating Enzyme Acetyl CoA synthetase. ACS Infect Dis 2016; 2:268-80; PMID:27088128; http://dx.doi.org/10.1021/acsinfecdis.5b00134
  • Abadio AKR, Kioshima ES, Teixeira MM, Martins NF, Maigret B, Felipe MSS. Comparative genomics allowed the identification of drug targets against human fungal pathogens. BMC Genomics 2011; 12:75; PMID:21272313; http://dx.doi.org/10.1186/1471-2164-12-75
  • Abadio AKR, Kioshima ES, Leroux V, Martins NF, Maigret B, Felipe MSS. Identification of new antifungal compounds targeting thioredoxin reductase of paracoccidioides genus. PLoS One 2015; 10:e0142926; PMID:26569405; http://dx.doi.org/10.1371/journal.pone.0142926
  • Edwards JA, Kemski MM, Rappleye CA. Identification of an aminothiazole with antifungal activity against intracellular Histoplasma capsulatum. Antimicrob Agents Chemother 2013; 57:4349-59; PMID:23817367; http://dx.doi.org/10.1128/AAC.00459-13
  • Isono K, Asahi K, Suzuki S. Studies on polyoxins, antifungal antibiotics. 13. The structure of polyoxins. J Am Chem Soc 1969; 91:7490-505; PMID:5358618; http://dx.doi.org/10.1021/ja01054a045
  • Endo A, Misato T. Polyoxin D, a competitive inhibitor of UDP-N-acetylglucosamine: chitin N-acetylglucosaminyltransferase in Neurospora crassa. Biochem Biophys Res Commun 1969; 37:718-22; PMID:5353899; http://dx.doi.org/10.1016/0006-291X(69)90870-5
  • Garfoot AL, Shen Q, Wüthrich M, Klein BS, Rappleye CA. The Eng1 β-Glucanase Enhances Histoplasma Virulence by Reducing β-Glucan Exposure. MBio 2016; 7:e01388-15; PMID:27094334; http://dx.doi.org/10.1128/mBio.01388-15
  • Goldberg J, Connolly P, Schnizlein-Bick C, Durkin M, Kohler S, Smedema M, Brizendine E, Hector R, Wheat J. Comparison of nikkomycin Z with amphotericin B and itraconazole for treatment of histoplasmosis in a murine model. Antimicrob Agents Chemother 2000; 44:1624-9; PMID:10817719; http://dx.doi.org/10.1128/AAC.44.6.1624-1629.2000
  • Hector RF, Zimmer BL, Pappagianis D. Evaluation of nikkomycins X and Z in murine models of coccidioidomycosis, histoplasmosis, and blastomycosis. Antimicrob Agents Chemother 1990; 34:587-93; PMID:2344165; http://dx.doi.org/10.1128/AAC.34.4.587
  • Graybill JR, Najvar LK, Bocanegra R, Hector RF, Luther MF. Efficacy of nikkomycin Z in the treatment of murine histoplasmosis. Antimicrob Agents Chemother 1998; 42:2371-4; PMID:9736565
  • Clemons KV, Stevens DA. Efficacy of nikkomycin Z against experimental pulmonary blastomycosis. Antimicrob Agents Chemother 1997; 41:2026-8; PMID:9303408
  • Kriesel JD, Sutton DA, Schulman S, Fothergill AW, Rinaldi MG. Persistent pulmonary infection with an azole-resistant Coccidioides species. Med Mycol 2008; 46:607-10; PMID:18608910; http://dx.doi.org/10.1080/13693780802140923
  • Wang CY, Jerng JS, Ko JC, Lin MF, Hsiao CH, Lee LN, Hsueh PR, Kuo SH. Disseminated coccidioidomycosis. Emerging Infect Dis 2005; 11:177-9; PMID:15714666; http://dx.doi.org/10.3201/eid1101.040613
  • Wheat LJ, Connolly P, Smedema M, Brizendine E, Hafner R, AIDS Clinical Trials Group and the Mycoses Study Group of the National Institute of Allergy and Infectious Diseases. Emergence of resistance to fluconazole as a cause of failure during treatment of histoplasmosis in patients with acquired immunodeficiency disease syndrome. Clin Infect Dis 2001; 33:1910-3; PMID:11692303; http://dx.doi.org/10.1086/323781
  • Kathuria S, Singh PK, Meis JF, Chowdhary A. In vitro antifungal susceptibility profile and correlation of mycelial and yeast forms of molecularly characterized Histoplasma capsulatum strains from India. Antimicrob Agents Chemother 2014; 58:5613-6; PMID:24982084; http://dx.doi.org/10.1128/AAC.02973-14
  • Ramani R, Chaturvedi V. Antifungal susceptibility profiles of Coccidioides immitis and Coccidioides posadasii from endemic and non-endemic areas. Mycopathologia 2007; 163:315-9; PMID:17484074; http://dx.doi.org/10.1007/s11046-007-9018-7
  • Li RK, Ciblak MA, Nordoff N, Pasarell L, Warnock DW, McGinnis MR. In vitro activities of voriconazole, itraconazole, and amphotericin B against Blastomyces dermatitidis, Coccidioides immitis, and Histoplasma capsulatum. Antimicrob Agents Chemother 2000; 44:1734-6; PMID:10817743; http://dx.doi.org/10.1128/AAC.44.6.1734-1736.2000
  • Sugar AM, Liu XP. In vitro and in vivo activities of SCH 56592 against Blastomyces dermatitidis. Antimicrob Agents Chemother 1996; 40:1314-6; PMID:8723494
  • Cordeiro RA, Brilhante RSN, Rocha MFG, Fechine MAB, Costa AKF, Camargo ZP, Sidrim JJC. In vitro activities of caspofungin, amphotericin B and azoles against Coccidioides posadasii strains from Northeast, Brazil. Mycopathologia 2006; 161:21-6; PMID:16389480; http://dx.doi.org/10.1007/s11046-005-0177-0
  • Cruz RC, Werneck SMC, Oliveira CS, Santos PC, Soares BM, Santos DA, Cisalpino PS. Influence of different media, incubation times, and temperatures for determining the MICs of seven antifungal agents against Paracoccidioides brasiliensis by microdilution. J Clin Microbiol 2013; 51:436-43; PMID:23175254; http://dx.doi.org/10.1128/JCM.02231-12
  • Andreu CMF, León AM, Medina YE, Machín GM, Lancha MRP, Zaragozí MTI. In vitro sensitivity of Histoplasma capsulatum var. capsulatum to amphotericin B, ketoconazole, itroconazole and fluconazole. Rev Cubana Med Trop 2003; 55:76-82; PMID:15849960
  • Zhanel GG, Karlowsky JA, Harding GA, Balko TV, Zelenitsky SA, Friesen M, Kabani A, Turik M, Hoban DJ. In vitro activity of a new semisynthetic echinocandin, LY-303366, against systemic isolates of Candida species, Cryptococcus neoformans, Blastomyces dermatitidis, and Aspergillus species. Antimicrob Agents Chemother 1997; 41:863-5; PMID:9087508
  • González GM, Tijerina R, Najvar LK, Bocanegra R, Luther M, Rinaldi MG, Graybill JR. Correlation between antifungal susceptibilities of Coccidioides immitis in vitro and antifungal treatment with caspofungin in a mouse model. Antimicrob Agents Chemother 2001; 45:1854-9; http://dx.doi.org/10.1128/AAC.45.6.1854-1859.2001
  • Kohler S, Wheat LJ, Connolly P, Schnizlein-Bick C, Durkin M, Smedema M, Goldberg J, Brizendine E. Comparison of the echinocandin caspofungin with amphotericin B for treatment of histoplasmosis following pulmonary challenge in a murine model. Antimicrob Agents Chemother 2000; 44:1850-4; PMID:10858342; http://dx.doi.org/10.1128/AAC.44.7.1850-1854.2000
  • LeMonte AM, Washum KE, Smedema ML, Schnizlein-Bick C, Kohler SM, Wheat LJ. Amphotericin B combined with itraconazole or fluconazole for treatment of histoplasmosis. J Infect Dis 2000; 182:545-50; PMID:10915087; http://dx.doi.org/10.1086/315717
  • Chapman SW, Rogers PD, Rinaldi MG, Sullivan DC. Susceptibilities of clinical and laboratory isolates of Blastomyces dermatitidis to ketoconazole, itraconazole, and fluconazole. Antimicrob Agents Chemother 1998; 42:978-80; PMID:9559827
  • González GM, Fothergill AW, Sutton DA, Rinaldi MG, Loebenberg D. In vitro activities of new and established triazoles against opportunistic filamentous and dimorphic fungi. Med Mycol 2005; 43:281-4; http://dx.doi.org/10.1080/13693780500088416
  • Van Duin D, Casadevall A, Nosanchuk JD. Melanization of Cryptococcus neoformans and Histoplasma capsulatum reduces their susceptibilities to amphotericin B and caspofungin. Antimicrob Agents Chemother 2002; 46:3394-400; PMID:12384341; http://dx.doi.org/10.1128/AAC.46.11.3394-3400.2002
  • Wheat LJ, Connolly P, Smedema M, Durkin M, Brizendine E, Mann P, Patel R, McNicholas PM, Goldman M. Activity of newer triazoles against Histoplasma capsulatum from patients with AIDS who failed fluconazole. J Antimicrob Chemother 2006; 57:1235-9; PMID:16627592; http://dx.doi.org/10.1093/jac/dkl133

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.