Next Article in Journal
Hog1 Controls Lipids Homeostasis Upon Osmotic Stress in Candida albicans
Next Article in Special Issue
Replicative Aging in Pathogenic Fungi
Previous Article in Journal
Relationship between Species Richness, Biomass and Structure of Vegetation and Mycobiota along an Altitudinal Transect in the Polar Urals
Previous Article in Special Issue
Insights into the Multi-Azole Resistance Profile in Candida haemulonii Species Complex
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Coccidioidomycosis: Changing Concepts and Knowledge Gaps

Department of Infectious Diseases, Medicine and Immunobiology University of Arizona, 1501 North Campbell Avenue, Tucson, AZ 85724, USA
J. Fungi 2020, 6(4), 354; https://doi.org/10.3390/jof6040354
Submission received: 26 October 2020 / Revised: 2 December 2020 / Accepted: 3 December 2020 / Published: 10 December 2020
(This article belongs to the Special Issue Systemic and Emerging Mycoses)

Abstract

:
Although first described more than 120 years ago, much remains unknown about coccidioidomycosis. In this review, new information that has led to changing concepts will be reviewed and remaining gaps in our knowledge will be discussed. In particular, new ideas regarding ecology and epidemiology, problems and promises of diagnosis, controversies over management, and the possibility of a vaccine will be covered.

1. Introduction

As stated more than two decades ago, coccidioidomycosis is a regional disease of national importance [1]. Recent studies attest to the continued medical [2] and economic impact [3] of this mycosis in the United States as well as in the rest of the western hemisphere [4,5]. In this review, particular aspects of coccidioidomycosis will be discussed that emphasize changing ideas and critical knowledge gaps. These will include new concepts regarding ecology, changing epidemiology, and possible geographic spread; problems and challenges with diagnosis; issues with treatment; and the possibility of a protective vaccine. While reference to older works will be noted when appropriate, the purpose here will be to cite more recent publications.

2. Defining the Ecology and Changing Epidemiology of Coccidioidomycosis

The distribution of coccidioidomycosis in nature was inferred in the past using three sources: epidemiology of proven cases, population-based skin test surveys, and ecological studies [6]. From these data, a model of coccidioidal infection was derived in which it was presumed that the fungus inhabits a soil niche and, either through direct exposure or due to air-borne spread, susceptible individuals acquire infection, usually through a respiratory route. While this model appears to be generally true, it is incomplete. With advances in molecular genomics, our understanding of the ecology and epidemiology of coccidioidomycosis is now changing and expanding. This began when it was established that Coccidioides consisted of two distinct species, C. immitis and C. posadasii [7]. Since then, the construction of phylogenetic trees has demonstrated that these species have distinct but overlapping geographic ranges [8,9] and have recently hybridized and exchanged genes [10]. Molecular genomics has now become refined enough to distinguish where a coccidioidal infection was geographically acquired [11].
However, the precise environments occupied by Coccidioides are still not adequately described. While it is clear that the fungus has a predilection for certain soils [12], it has been notoriously difficult to isolate using traditional culture methods [9] and its particular environmental niche remains unclear. Recently, the idea that small, soil-dwelling animals might serve as the primary environmental reservoir for Coccidioides [13], rather than the soil itself, has been revived. Taylor and Barker [14] have proposed that Coccidioides is not a saprophyte whose life cycle is a dead-end within the infected mammalian host, but rather an endozoan, able to live in the granulomata of infected mammals and, upon death of the host, use local nutrients released in the surrounding soil for nourishment. This concept is supported by genomic analysis of Coccidioides demonstrating, when compared to other filamentous Ascomycetes, that there are expansions of protease families and that the numbers of plant cell-wall degrading enzymes are reduced compared to fungi known to decay plant matter [15]. This hypothesis has been strengthened by experimental data. DNA extracted from 465 soil samples from five Arizona locations was subjected to real-time qPCR. Of these, 105 samples (23%) were positive for Coccidioides and 95 of these were from animal burrows [16]. These results suggest that Coccidioides is not extant in the soil environment, but rather resides in focal pockets in association with small animals and their burrows. These results are supported by studies in the northern Baja region of Mexico that have used both molecular detection of Coccidioides in the soil and antibody detection of small rodents [17,18]. Moreover, this hypothesis fits with the concept that certain fungi, including members of the order Onygenales to which Coccidioides belongs, may be part of the normal human lung microbiome [19].
The conditions for air-borne spread and respiratory acquisition have also remained elusive. It has been estimated that a single arthroconidium is sufficient to cause infection in an animal model [20]. However, there is a clinical sense that exposure to higher inocula results in more severe respiratory disease [21] and that the risk of extrapulmonary dissemination may be increased with exposure to higher respiratory inocula [22]. The median time of living in the coccidioidal endemic area in Arizona at the time of serological conversion has been estimated to be 6.5 years and appears to increase linearly with time [23]. However, the risk of infection also appears to be stochastic and dependent on specific occurrences, such as dust storms and earthquakes [24]. These data imply that a variety of events, many currently undefined, may play important roles in predicting whether an individual acquires coccidioidal infection.
In the past, assessing the air-borne spread of Coccidioides has been difficult. However, genomics may be improving this. In one recent study, air was sampled and filtered from 21 sites in the Phoenix metropolitan area on a specific day after a dust storm as well as over a 45-day period during the fall. DNA was extracted from the filters and subjected to nested qPCR [25]. Although prior evidence has suggested that dust storms are associated with outbreaks of coccidioidomycosis [26], there was actually a decrease in positive filter samples from sites on the days immediately after the dust storm compared to before. Moreover, there were both temporal and geographic differences in filter positivity during the 45-day sample period. Clearly, more such studies are needed and a linkage to human cases will be required to flesh out associations. However, the ability to consistently detect Coccidioides in the air is a critical first step.
Finally, in recent years, environmental sites containing Coccidioides that are well outside of the traditionally recognized coccidioidal endemic zones have been associated with outbreaks of coccidioidomycosis. These include an ancient Amerindian location in northeast Utah [27] and an area of eastern Washington state [28]. Moreover, the areas where Coccidioides might reside appears to be much larger than the sites already identified [29]. Because the organisms at these sites have likely been there for millennia, one must conclude that these outbreaks are associated with some recent alteration, such as climate change or human population expansion. Recently, Gorris and her colleagues have suggested [30], using a county-level database of cases and climatic and environmental information, that the endemic region of coccidioidomycosis is likely to extend beyond currently recognized boundaries and that it will increase within the known endemic regions as the climate changes.

3. Issues with Diagnosis

While coccidioidomycosis may be diagnosed in a variety of ways, serology has by far remained the most common method used. Because most clinical laboratories have the appropriate platform and because the results are generally available within 24 h, enzyme immunoassays (EIAs) are most frequently employed. However, there is no established standard by which the several available commercial EIAs can be compared and all use proprietary antigens that are not described [31]. This had led to a certain amount of uncertainty with regard to their meaning and interpretation in certain clinical situations.
EIA coccidioidal antibody testing is particularly useful among patients who present with a typical clinical syndrome of coccidioidomycosis within the endemic area [32]. In such instances, when both the IgM and IgM assays are positive, it is highly likely that the patient has primary pulmonary coccidioidomycosis. However, these tests are less useful in other situations, such as screening immunocompromised patients [33]. This was recently emphasized in a retrospective review of rheumatology patients in the endemic area who were routinely screened for coccidioidal antibody [31]. In that study, the finding of a positive coccidioidal serology in the absence of symptoms did not predict clinical illness.
Recently, McHardy and colleagues reviewed their experience at the University of California at Davis with the coccidioidal complement fixation (CF) test in the age of antifungal therapy [34]. From their large database, they found that titers generally reflected clinical illness, with the lowest ones found in those with uncomplicated pulmonary disease and higher levels with disseminated extrathoracic illness. While they did not find a specific titer predictive of disseminated disease, a titer ≥ 1:32 suggested a complicated clinical course. They also noted that the change in CF titers was slow, with a decline by one dilution every 90 days for those with uncomplicated pulmonary disease and longer for other patients. These data suggest that the test should be ordered at appropriately long intervals to be used as a measure of improvement.
Delays in diagnosis of coccidioidomycosis have been shown to be costly [35] and more rapid diagnostic tests are needed. Recently, a lateral flow antibody assay (LFA) that could allow bedside performance with results in under one hour has been developed [36]. Unfortunately, a recent assessment suggests that it may have an unacceptably low sensitivity when compared to EIA [37]. Direct assays of Coccidioides are also available. The measurement of β-1,3-d-glucan (BDG) is a non-specific fungal assay that is now widely available in clinical laboratories. Unfortunately, in one study, it had a sensitivity of only 19% at a cut-off level of ≥80 pg/mL among patients with acute pulmonary coccidioidomycosis [38]. More promising is PCR assessment. These assays have been shown to be rapid, specific, and recently capable of being performed in clinical microbiology laboratories [39,40]. Although the overall sensitivity, particularly compared to serology, is not known, these tests could become very useful if they were to become more generally available.
There has been renewed interest in assessing the cellular immune response to coccidioidomycosis as both a diagnostic and a prognostic tool. In a study of a reformulated spherule-based reagent [41], 52 of 53 subjects (98%) with acute pulmonary coccidioidomycosis demonstrated ≥5 mm of induration 48 h after intradermal injection. Based on this, the test was approved in 2014 for use in the United States. A subsequent study of 36,789 inmates in a prison system demonstrated an 8.6% positivity rate that correlated with residence in the endemic region [42]. The results were used to restrict inmates with negative tests from incarceration at two prisons with the highest coccidioidomycosis rates. The use of the test for prognostic purposes is less clear. Two recent studies of patients followed for various types of coccidioidomycosis have suggested that expression of delayed type hypersensitivity reactions following skin testing may not always correlate with clinical expression of disease or be prognostically helpful [43,44].
Intradermal skin testing has several drawbacks. It requires skill and training both in the placement and in the interpretation of the result and it requires an additional visit 48 h after placement to read the response. In addition, there is a predictable incidence of adverse events, nearly 5% among those in the prison study [42]. Perhaps because of these issues, the test has not been extensively adopted for use in the United States since its introduction [45]. There has been interest in developing a whole blood assay measuring cytokine release after ex vivo antigen exposure. This method has been shown to correlate with skin test dermal hypersensitivity [46] and has been studied in several experimental models [47,48,49]. However, at this time, no commercial assay is available.
Several tests are on the horizon. Recently, Peng and colleagues [50] reported using recombinant CST1, a fungal chitinase that is the antigen for the IgG antibody response [51], to perform quantitative coccidioidal titers using an enzyme-linked system. They were able to identify a 200-amino acid segment of the gene product that specifically bound antibody. The assay correlated well with the standard CF assay but with the potential advantage that the test could be performed by many clinical laboratories in much less time than the current method.
Coccidioides produces a variety of volatile organic compounds (VOC) that can be detected using solid phase microextraction (SPME) and comprehensive two-dimensional gas chromatography time-of-flight mass spectrometry (GC × GC-TOFMS). The number of these metabolites was found to be increased in a wild-type strain of C. posadasii compared to an attenuated mutant strain [52]. Detecting such gas-phase metabolites could be used as a potentially rapid diagnostic tool through breath analysis. Preliminary data have suggested the feasibility of this in an experimental model studying multiple strains of C. posadasii and C. immitis [53].
Finally, using a targeted liquid chromatography-tandem mass spectrometry-based (LC-MS/MS) metabolic profiling approach, 207 plasma and 231 urinary coccidioidal metabolites were reliably detected in high abundance in samples from 48 patients with clinical coccidioidomycosis compared to 99 individuals without that diagnosis [54]. Statistical modeling allowed the identification of three significantly altered plasma metabolites and nine urine metabolites in patients with coccidioidomycosis. Constructing receiver–operator curves demonstrated that the plasma metabolites had a sensitivity of >94% and a specificity of >97%. Because LC-MS/MS is now available in many clinical laboratories, this too could become a rapid assay for the diagnosis of coccidioidomycosis.

4. Treatment: When and What

Prior to the development of antifungal drugs in the 1950s, it was demonstrated that fully 60% of individuals who acquired coccidioidal infection did so without symptoms and the vast majority of those with symptoms resolved their clinical disease without sequelae and with apparent long-lived immunity [55]. The relative benignity of pulmonary compared to extrathoracic disseminated coccidioidomycosis in the age before antifungals was recently demonstrated in a retrospective review of 531 patients followed until 1966 as part of the VA-Armed Forces Cooperative Study [56]. In that study, the all-cause mortality of pulmonary coccidioidomycosis was 5.4% over 30 years, significantly lower than 30% in those with non-meningeal disseminated disease and 96% in those with meningeal coccidioidomycosis.
With the development of oral triazole antifungal therapy in the 1990s, treatment of symptomatic primary pulmonary coccidioidomycosis became a common practice, despite the lack of controlled trials. Two retrospective cohort studies have suggested that there may be no clear benefit from antifungal therapy for such patients. In the first [57], 43 patients with primary pulmonary coccidioidomycosis were followed for a median 286 days. Among 16 who received triazole antifungal therapy, at the discretion of the treating providers, two developed extrathoracic dissemination after treatment was discontinued, while all 36 who received no therapy had an uncomplicated outcome. Moreover, the rate of improvement was not different between the two groups. In the second study [58], 27 patients with primary pulmonary coccidioidomycosis were followed for 24 weeks. Twenty patients, again at the discretion of the providers, were prescribed antifungal therapy. As in the previous study, median times to improvement between the two groups were not different and one patient developed extrathoracic dissemination; that patient was in the treatment group. Because both of these studies were not controlled with regard to therapy, they were potentially biased. In an attempt to definitively answer whether early antifungal therapy leads to improved outcomes for those with primary pulmonary coccidioidomycosis, the National Institute of Allergy and Infectious Diseases (NIAID) sponsored a study that provided 42 days of fluconazole or placebo for patients living in the coccidioidal endemic region with presumed coccidioidal pneumonia (Clinical trials.gov identifier: NCT02663674). Unfortunately, this trial ended early due to lack of enrollment. For now, it seems that patients without underlying conditions and without evidence of severe infection may not require antifungal therapy for uncomplicated primary pulmonary coccidioidomycosis.
A second important therapy question is what antifungal agent is best to use for coccidioidomycosis. There has be a consistent deference toward using fluconazole in this situation, both because this was the triazole antifungal initially studied in coccidioidomycosis and because it was perceived to have the most benign toxicity profile. However, in fact, there are data to challenge this view. First, in early non-comparative studies of chronic coccidioidomycosis, relapses were less frequent in those who received itraconazole compared to fluconazole [59,60]. In the only randomized, double-blind comparative trial of fluconazole and itraconazole for chronic non-meningeal coccidioidomycosis, the response rate to itraconazole was significantly higher than fluconazole in those with skeletal infections [61]. Moreover, when patients fail fluconazole, a switch to another triazole is frequently effective [62]. The preferred therapy for two related fungal infections, histoplasmosis and blastomycosis, is not fluconazole but rather itraconazole. Finally, two in vitro susceptibility studies have demonstrated that, of all the available triazole antifungals, fluconazole has by far the highest minimum inhibitory concentrations (MICs) against Coccidioides [63,64] (Table 1). Unfortunately, there are no recent comparative trials of triazole antifungals for coccidioidomycosis. These are now urgently needed. In the meantime, a reevaluation of the deference toward the use of fluconazole for coccidioidomycosis should be reconsidered.
The triazole antifungals have distinct toxicities inherently related to their mechanism of action. All exert their antifungal effect by preventing the conversion of lanosterol to ergosterol by inhibiting 14-α-demethylase [65], a reaction dependent upon CYP51, which is also present in humans [66]. In addition, cross-inhibition of several human CYP-dependent enzymes, particularly CYP3A4, 2C9, and 2C19, is responsible for many of the clinical side effects and drug–drug interactions. The latter are of particular concern, as many pharmacological agents depend on these systems for their metabolism. Inhibition of these metabolic pathways by triazole antifungals can result in significant toxicity [67]. Particularly common is the increase in coagulation seen when these agents are combined with warfarin and the increased in serum levels and toxicity that occur when combined with the calcineurin inhibitors cyclosporine and tacrolimus. Although fluconazole has been considered the least toxic of the triazole antifungals, a long-term use has been shown to result in adverse effects in the majority of patients. Among the most common are xerosis, alopecia, and fatigue [68]. The triazole antifungals have also been associated with teratogenicity, an effect that appears to be directly related to inhibition of human CYP51. They should be avoided in women seeking to become pregnant and during the first trimester of pregnancy [69]. There are also unique toxicities. Voriconazole, which is trifluorinated, has been associated with periostitis due to elevated fluoride levels [70,71]. Recently, the delayed-release formulation of posaconazole has been associated with the syndrome of pseudoaldosteronism [72]. Patients have presented with severe hypertension and hypokalemia often associated with elevated drug levels that resolves with stopping or reducing the dosage of the medication. The mechanism appears related to the inhibition of 11β-hydroxysteroid activity [73].

5. Antifungals on the Horizon

Nikkomycin Z (Table 2) is a competitive inhibitor of chitin synthase because of its structural similarity to uridine diphosphate-N-acetylglucosamine, a monosaccharide building block of fungal chitin. The target is lacking in human hosts [74]. Hector and colleagues [75] demonstrated significant in vitro activity against both Coccidioides and Blastomyces, but less activity against yeast and none against Aspergillus species. Doses of 20 and 50 mg/kg were completely protective in a murine intranasal coccidioidal infection model. More importantly, treatment with the 50 mg/kg dose for six days eradicated nearly all of the fungus from the lungs of treated animals; a single colony of Coccidioides was recovered from one of eight mice infected and treated. This suggests that it is fungicidal and potentially curative. Li and Rinaldi subsequently demonstrated that combining nikkomycin Z with either fluconazole or itraconazole resulted in synergistic in vitro activity against a variety of fungal pathogens [76]. A pharmacological study in humans revealed that a single oral dose was well tolerated and resulted in no toxicity with therapeutic bioavailability and a half-life of 1–2 h [77]. Shubitz and colleagues completed treatment of nine canines with coccidioidomycosis in an open-label study at doses of either 250 mg or 500 mg daily for an average of three months. Most had been previously treated with fluconazole. Overall, seven of the dogs improved. No toxicity was observed and the twice-daily therapy resulted in acceptable pharmacokinetics [78]. Subsequently, it was shown that a dose of 80 mg/kg daily given in two doses for three weeks in mice resulted in optimal clearance of Coccidioides from the lungs with acceptable pharmacokinetics that could be achieved in humans [79]. Although nikkomycin Z has been granted qualified infectious diseases product (QIDP) designation by the Food and Drug Administration, at this time, it is not being studied further in humans until more is manufactured.
Tetrazoles are a new class of antifungals that have specific, selective activity against fungal but not mammalian 14-α-demethylase (CYP51) [74]. Because of this, these agents avoid many of the CYP-related toxicities associated with triazole antifungals detailed above. One of these, VT-1598 (Table 2), has been studied specifically against Coccidioides. Using an in vitro model, VT-1598 had MICs against clinical isolates of Coccidioides between 0.06 and 0.50 µg/mL, comparable to posaconazole and far below that of fluconazole [80]. In a murine model of coccidioidal meningitis, VT-1598 at 20 mg/kg, beginning two days after inoculation with either the Silveira strain of C. posadasii or a clinical isolate of C. immitis, resulted in protection from death that appeared to be superior to fluconazole at 25 mg/kg. Plasma levels remained in the inhibitory range two days after the last treatment [81]. Like nikkomycin Z, VT-1578 has QIDP designation and human studies are being planned.
Olorofim (Table 2) is a member of a new class of antifungals, the orotomides, that reversibly inhibits fungal pyrimidine biosynthesis through the enzyme dihydroorotate dehydrogenase (DHODH) with little inhibition of the human enzyme [82,83]. It has demonstrated in vitro activity against Coccidioides [80,82]. In a murine model of coccidioidal meningitis, doses of 10 and 20 mg/kg appeared to confer a survival advantage over fluconazole at 25 mg/kg, although there were still deaths [84]. An intravenous formulation using a β-hydroxypropyl-cyclodextrin vehicle has been studied in human volunteers with a half-life between 20 and 30 h. An oral formulation is also being studied [83]. Human studies for coccidioidomycosis are being planned.

6. Vaccines

As pointed out by Kirkland, immunization for coccidioidomycosis seems feasible since second infections appear to be extraordinarily rare and initial infection appears to confer life-long immunity [85]. Barnato and colleagues in 2001 proposed an economic case for the development of a vaccine using a decision model [86]. They estimated that adult vaccination in the coccidioidal endemic regions would result in a savings of $62,000 per quality adjusted life year compared to no vaccination and would result in 11 fewer deaths and save $3 million annually. Current data appear to be even more in favor of vaccination. In California during 2017, the estimated direct and indirect costs for 7466 patients was nearly $700 million [3]. In Arizona, the costs of managing delays in diagnoses were nearly $600,000 in just under a three-year period [35].
Because of this, there has been renewed interest in a coccidioidal vaccine since the publication in 1993 of the inconclusive results of a formaldehyde-killed thimerosal-preserved spherule vaccine [87,88]. For this renewed effort, the goals of vaccination would be multiple. A vaccine should provide protection against primary acute infection, prevent extrathoracic dissemination, and protection should persist for a prolonged if not life-long period and not wane during immunosuppression, and the vaccine should be safe and well-tolerated.
Two approaches have been taken. In the first, a live mutant strain of C. posadasii has been created by deleting the CPS1 gene, a fungal virulence factor. The resultant Δcsp1 strain demonstrates slower growth and smaller spherules than wild-type when grown under in vitro conditions. Moreover, the Δcsp1 strain was found to be avirulent in mice and, when used as a vaccine, resulted in protection from wild-type coccidioidal infection in susceptible mouse strains [89]. This has resulted in an NIAID sponsored study to develop the vaccine in a canine model. Results for this study are currently pending, but if successful, it could serve as a bridge for the development of a human vaccine.
The second direction is to develop a sub-unit plus adjuvant vaccine using antigens that have induced protection in mouse models of coccidioidomycosis. In this system, a construct using three C. posadasii antigens, Ag2-PRA, Cs-Ag, and Pmp-1, collectively called rCpa1, have been attached to a five-peptide sequence containing human T-cell epitopes that allows for binding to human HLA Class II molecules. This is then incorporated into glucan–chitin particles derived from Rhodotorula mucilaginous, which acts as the adjuvant. Preliminary studies in mice have shown protection against strains of both C. immitis and C. posadasii [90], stimulation of the Th1/Th17 pathway [91], and activation of the CARD9 dectin-1 and dectin-2 signaling pathways [92]. Finally, no significant toxicity has been seen in a human liver cell line (HepG2) (personal communication, C-Y Hung). We will have to await further results from both these endeavors, but it is likely that a vaccine for coccidioidomycosis will be developed for humans in the foreseeable future.

Funding

This research received no external funding.

Conflicts of Interest

The author declares no conflict of interest.

References

  1. Galgiani, J.N. Coccidioidomycosis: A regional disease of national importance. Rethinking approaches for control. Ann. Intern. Med. 1999, 130, 293–300. [Google Scholar] [CrossRef]
  2. McCotter, O.Z.; Benedict, K.; Engelthaler, D.M.; Komatsu, K.; Lucas, K.D.; Mohle-Boetani, J.C.; Oltean, H.; Vugia, D.; Chiller, T.M.; Cooksey, G.L.S.; et al. Update on the Epidemiology of coccidioidomycosis in the United States. Med. Mycol. 2019, 57, S30–S40. [Google Scholar] [CrossRef]
  3. Wilson, L.; Ting, J.; Lin, H.; Shah, R.; MacLean, M.; Peterson, M.W.; Stockamp, N.; Libke, R.; Brown, P. The Rise of Valley Fever: Prevalence and Cost Burden of Coccidioidomycosis Infection in California. Int. J. Environ. Res. Public Health 2019, 16, 1113. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Laniado-Laborín, R.; Arathoon, E.G.; Canteros, C.; Muñiz-Salazar, R.; Rendon, A. Coccidioidomycosis in Latin America. Med. Mycol. 2019, 57, S46–S55. [Google Scholar] [CrossRef]
  5. Ashraf, N.; Kubat, R.C.; Poplin, V.; Adenis, A.; Denning, D.W.; Wright, L.; McCotter, O.; Schwartz, I.S.; Jackson, B.R.; Chiller, T.; et al. Re-drawing the Maps for Endemic Mycoses. Mycopathologia 2020, 185, 843–865. [Google Scholar]
  6. Drutz, J.D.; Catanzaro, A. Coccidioidomycosis: Part I. Am. Rev. Respir. Dis. 1978, 117, 559–585. [Google Scholar]
  7. Fisher, M.C.; Koenig, G.L.; White, T.J.; Taylor, J.W. Molecular and Phenotypic Description of Coccidioides posadasii sp. nov., Previously Recognized as the Non-California Population of Coccidioides immitis. Mycologia 2002, 94, 73–84. [Google Scholar] [CrossRef] [Green Version]
  8. Engelthaler, D.M.; Roe, C.C.; Hepp, C.M.; Teixeira, M.; Driebe, E.M.; Schupp, J.M.; Gade, L.; Waddell, V.; Komatsu, K.; Arathoon, E.; et al. Local Population Structure and Patterns of Western Hemisphere Dispersal for Coccidioides spp. the Fungal Cause of Valley Fever. MBio 2016, 7, e00550. [Google Scholar] [CrossRef] [Green Version]
  9. Barker, B.M.; Litvintseva, A.P.; Riquelme, M.; Vargas-Gastélum, L. Coccidioides ecology and genomics. Med. Mycol. 2019, 57 (Suppl. S1), S21–S29. [Google Scholar] [CrossRef]
  10. Neafsey, D.E.; Barker, B.M.; Sharpton, T.J.; Stajich, J.E.; Park, D.J.; Whiston, E.; Hung, C.-Y.; Mcmahan, C.; White, J.; Sykes, S.; et al. Population genomic sequencing of Coccidioides fungi reveals recent hybridization and transposon control. Genome Res. 2010, 20, 938–946. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  11. Barker, B.M.; Rajan, S.; Teixeira, M.D.M.; Sewnarine, M.; Roe, C.; Engelthaler, D.M.; Galgiani, J.N. Coccidioidal Meningitis in New York Traced to Texas by Fungal Genomic Analysis. Clin. Infect. Dis. 2019, 69, 1060–1062. [Google Scholar] [CrossRef] [PubMed]
  12. Fisher, F.S.; Bultman, M.W.; Johnson, S.M.; Pappagianis, D.; Zaborsky, E. Coccidioidesniches and habitat parameters in the southwestern United States: A matter of scale. Ann. N. Y. Acad. Sci. 2007, 1111, 47–72. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Emmons, C.W. Coccidioidomycosis in Wild Rodents. A Method of Determining the Extent of Endemic Areas. Public Health Rep. (1896–1970) 1943, 58, 1–5. [Google Scholar] [CrossRef]
  14. Taylor, J.W.; Barker, B.M. The endozoan, small-mammal reservoir hypothesis and the life cycle of Coccidioides species. Med. Mycol. 2019, 57, S16–S20. [Google Scholar] [CrossRef]
  15. Sharpton, T.J.; Stajich, J.E.; Rounsley, S.D.; Gardner, M.J.; Wortman, J.R.; Jordar, V.S.; Maiti, R.; Kodira, C.D.; Neafsey, D.E.; Zeng, Q.; et al. Comparative genomic analyses of the human fungal pathogens Coccidioides and their relatives. Genome Res. 2009, 19, 1722–1731. [Google Scholar] [CrossRef] [Green Version]
  16. Kollath, D.R.; Teixeira, M.M.; Funke, A.; Miller, K.J.; Barker, B.M. Investigating the Role of Animal Burrows on the Ecology and Distribution of Coccidioides spp. in Arizona Soils. Mycopathol. 2019, 185, 1–15. [Google Scholar] [CrossRef]
  17. Baptista-Rosas, R.C.; Catalán-Dibene, J.; Romero-Olivares, A.L.; Hinojosa, A.; Cavazos, T.; Riquelme, M. Molecular detection of Coccidioides spp. from environmental samples in Baja California: Linking Valley Fever to soil and climate conditions. Fungal Ecol. 2012, 5, 177–190. [Google Scholar] [CrossRef]
  18. Catalán-Dibene, J.; Johnson, S.M.; Eaton, R.; Romero-Olivares, A.L.; Baptista-Rosas, R.C.; Pappagianis, D.; Riquelme, M. Detection of coccidioidal antibodies in serum of a small rodent community in Baja California, Mexico. Fungal Biol. 2014, 118, 330–339. [Google Scholar] [CrossRef]
  19. Hamm, P.S.; Taylor, J.W.; Cook, J.A.; Natvig, D.O. Decades-old studies of fungi associated with mammalian lungs and modern DNA sequencing approaches help define the nature of the lung mycobiome. PLoS Pathog. 2020, 16, e1008684. [Google Scholar] [CrossRef]
  20. Shubitz, L.; Peng, T.; Perrill, R.; Simons, J.; Orsborn, K.; Galgiani, J.N. Protection of Mice against Coccidioides immitis Intranasal Infection by Vaccination with Recombinant Antigen 2/PRA. Infect. Immun. 2002, 70, 3287–3289. [Google Scholar] [CrossRef] [Green Version]
  21. Larsen, R.; Jacobson, J.; Morris, A.H.; Benowitz, B. Acute respiratory failure caused by primary pulmonary coccidioidomycosis. Two case reports and a review of the literature. Am. Rev. Respir. Dis. 1985, 131, 797–799. [Google Scholar] [PubMed]
  22. Werner, S.B.; Pappagianis, D. Coccidioidomycosis in Northern California. An outbreak among archeology students near Red Bluff. Calif. Med. 1973, 119, 16–20. [Google Scholar] [PubMed]
  23. Leake, J.A.D.; Mosley, D.G.; England, B.; Graham, J.V.; Plikaytis, B.D.; Ampel, N.M.; Perkins, B.A.; Hajjeh, R.A. Risk Factors for Acute Symptomatic Coccidioidomycosis among Elderly Persons in Arizona, 1996–1997. J. Infect. Dis. 2000, 181, 1435–1440. [Google Scholar] [CrossRef] [PubMed]
  24. Kollath, D.R.; Miller, K.J.; Barker, B.M. The mysterious desert dwellers: Coccidioides immitis and Coccidioides posadasii, causative fungal agents of coccidioidomycosis. Virulence 2019, 10, 222–233. [Google Scholar] [CrossRef] [Green Version]
  25. Gade, L.; McCotter, O.Z.; Bowers, J.R.; Waddell, V.; Brady, S.; Carvajal, J.; Sunenshine, R.; Komatsu, K.K.; Engelthaler, D.M.; Chiller, T.; et al. The detection of Coccidioides from ambient air in Phoenix, Arizona: Evidence of uneven distribution and seasonality. Med. Mycol. 2019, 58, 552–559. [Google Scholar] [CrossRef]
  26. Pappagianis, D.; Einstein, H. Tempest from Tehachapi takes toll or Coccidioides conveyed aloft and afar. West. J. Med. 1978, 129, 527–530. [Google Scholar]
  27. Johnson, S.M.; Carlson, E.L.; Fisher, F.S.; Pappagianis, D. Demonstration of Coccidioides immitis and Coccidioides posadasii DNA in soil samples collected from Dinosaur National Monument, Utah. Med. Mycol. 2014, 52, 610–617. [Google Scholar] [CrossRef] [Green Version]
  28. Litvintseva, A.P.; Marsden-Haug, N.; Hurst, S.; Hill, H.; Gade, L.; Driebe, E.M.; Ralston, C.; Roe, C.; Barker, B.M.; Goldoft, M.; et al. Valley Fever: Finding New Places for an Old Disease: Coccidioides immitis Found in Washington State Soil Associated with Recent Human Infection. Clin. Infect. Dis. 2014, 60, e1–e3. [Google Scholar] [CrossRef] [Green Version]
  29. Oltean, H.N.; Springer, M.; Bowers, J.R.; Barnes, R.; Reid, G.; Valentine, M.; Engelthaler, D.M.; Toda, M.; McCotter, O.Z. Suspected Locally Acquired Coccidioidomycosis in Human, Spokane, Washington, USA. Emerg. Infect. Dis. 2020, 26, 606–609. [Google Scholar] [CrossRef]
  30. Gorris, M.E.; Treseder, K.K.; Zender, C.; Randerson, J.T. Expansion of Coccidioidomycosis Endemic Regions in the United States in Response to Climate Change. GeoHealth 2019, 3, 308–327. [Google Scholar] [CrossRef] [Green Version]
  31. Mirbod-Donovan, F.; Zangeneh, T.T.; Malo, J.; Galgiani, J.N. Top Questions in the Diagnosis and Treatment of Coccidioidomycosis. Open Forum Infect. Dis. 2017, 4, 197. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Blair, J.E.; Mendoza, N.; Force, S.; Chang, Y.-H.H.; Grys, T.E. Clinical Specificity of the Enzyme Immunoassay Test for Coccidioidomycosis Varies According to the Reason for Its Performance. Clin. Vaccine Immunol. 2012, 20, 95–98. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Bilal, J.; Kollampare, S.; Bode, B.; Lisse, J.R.; Hoover, S.E.; Sudano, D.; Ampel, N.M. Management of asymptomatic coccidioidomycosis in patients with rheumatic diseases. Rheumatol. Int. 2019, 39, 1257–1262. [Google Scholar] [CrossRef]
  34. McHardy, I.H.; Dinh, B.-T.N.; Waldman, S.; Stewart, E.; Bays, D.; Pappagianis, D.; Thompson, G.R. Coccidioidomycosis Complement Fixation Titer Trends in the Age of Antifungals. J. Clin. Microbiol. 2018, 56. [Google Scholar] [CrossRef] [Green Version]
  35. Donovan, F.M.; Wightman, P.; Zong, Y.; Gabe, L.; Majeed, A.; Ynosencio, T.; Bedrick, E.J.; Galgiani, J.N. Delays in Coccidioidomycosis Diagnosis and Associated Healthcare Utilization, Tucson, Arizona, USA. Emerg. Infect. Dis. 2019, 25, 1745–1747. [Google Scholar] [CrossRef] [Green Version]
  36. Maddox, S.; Doherty, B.; Pelfrey, J.; Thompson, G.; Bauman, S. Rapid detection of anti-coccidioides antibodies using the sona™ Coccidioides Ab lateral flow assay. In Proceedings of the 7th International Conference on Coccidioidomycosis, Stanford, CA, USA, 10–13 August 2017. [Google Scholar]
  37. Donovan, F.M.; Ramadan, F.A.; Khan, S.A.; Bhaskara, A.; Lainhart, W.D.; Narang, A.T.; Mosier, J.M.; Ellingson, K.D.; Bedrick, E.J.; Saubolle, M.A.; et al. Comparison of a Novel Rapid Lateral Flow Assay to Enzyme Immunoassay Results for Early Diagnosis of Coccidioidomycosis. Clin. Infect. Dis. 2020. [Google Scholar] [CrossRef]
  38. Thompson, G.R., 3rd; Bays, D.J.; Johnson, S.M.; Cohen, S.H.; Pappagianis, D.; Finkelman, M.A. Serum (1->3)-beta-D-glucan measurement in coccidioidomycosis. J. Clin. Microbiol. 2012, 50, 3060–3062. [Google Scholar] [CrossRef] [Green Version]
  39. Saubolle, M.A.; Wojack, B.R.; Wertheimer, A.M.; Fuayagem, A.Z.; Young, S.; Koeneman, B.A. Multicenter Clinical Validation of a Cartridge-Based Real-Time PCR System for Detection of Coccidioides spp. in Lower Respiratory Specimens. J. Clin. Microbiol. 2017, 56, e01277-17. [Google Scholar] [CrossRef] [Green Version]
  40. Dizon, D.; Mitchell, M.; Dizon, B.; Libke, R.; Peterson, M.W. The utility of real-time polymerase chain reaction in detecting Coccidioides immitis among clinical specimens in the Central California San Joaquin Valley. Med. Mycol. 2019, 57, 688–693. [Google Scholar] [CrossRef]
  41. Johnson, R.; Kernerman, S.M.; Sawtelle, B.G.; Rastogi, S.C.; Nielsen, H.S.; Ampel, N.M. A Reformulated Spherule-Derived Coccidioidin (Spherusol) to Detect Delayed-Type Hypersensitivity in Coccidioidomycosis. Mycopathologia 2012, 174, 353–358. [Google Scholar] [CrossRef]
  42. Wheeler, C.; Lucas, K.D.; Derado, G.; McCotter, O.; Tharratt, R.S.; Chiller, T.; Mohle-Boetani, J.C. Risk Stratification with Coccidioidal Skin Test to Prevent Valley Fever Among Inmates, California, 2015. J. Correct. Health Care 2018, 24, 342–351. [Google Scholar] [CrossRef]
  43. Ampel, N.M.; Robey, I.; Nguyen, C.T. An Analysis of Skin Test Responses to Spherulin-Based Coccidioidin (Spherusol®) Among a Group of Subjects with Various Forms of Active Coccidioidomycosis. Mycopathologia 2019, 184, 533–538. [Google Scholar] [CrossRef]
  44. Mafi, N.; Murphy, C.B.; Girardo, M.E.; Blair, J.E. Coccidioides (spherulin) skin testing in patients with pulmonary coccidioidomycosis in an endemic regiondagger. Med. Mycol. 2020, 58, 626–631. [Google Scholar] [CrossRef]
  45. Benedict, K.; McCotter, O.Z.; Jackson, B.R. Coccidioidomycosis Skin Testing in a Commercially Insured Population, United States, 2014–2017. Emerg. Infect. Dis. 2020, 26, 619–621. [Google Scholar] [CrossRef] [Green Version]
  46. Ampel, N.M.; Hector, R.F.; Lindan, C.P.; Rutherford, G.W. An Archived Lot of Coccidioidin Induces Specific Coccidioidal Delayed-type Hypersensitivity and Correlates with in vitro Assays of Coccidioidal Cellular Immune Response. Mycopathologia 2006, 161, 67–72. [Google Scholar] [CrossRef]
  47. Nesbit, L.; Johnson, S.M.; Pappagianis, D.; Ampel, N.M. Polyfunctional T Lymphocytes Are in the Peripheral Blood of Donors Naturally Immune to Coccidioidomycosis and Are Not Induced by Dendritic Cells. Infect. Immun. 2010, 78, 309–315. [Google Scholar] [CrossRef] [Green Version]
  48. Nesbit, L.A.; Knox, K.S.; Nguyen, C.T.; Roesch, J.; Wheat, L.J.; Johnson, S.M.; Pappagianis, D.; Chavez, S.; Ampel, N.M. Immunological Characterization of Bronchoalveolar Lavage Fluid in Patients with Acute Pulmonary Coccidioidomycosis. J. Infect. Dis. 2013, 208, 857–863. [Google Scholar] [CrossRef] [Green Version]
  49. Ampel, N.M.; Nesbit, L.A.; Nguyen, C.T.; Chavez, S.; Knox, K.S.; Johnson, S.M.; Pappagianis, D. Cytokine Profiles from Antigen-Stimulated Whole-Blood Samples among Patients with Pulmonary or Nonmeningeal Disseminated Coccidioidomycosis. Clin. Vaccine Immunol. 2015, 22, 917–922. [Google Scholar] [CrossRef] [Green Version]
  50. Peng, T.; Zong, Y.; Johnson, M.D.; Menghani, S.V.; Lewis, M.L.; Galgiani, J.N. A quantitative enzyme-linked immunoassay (ELISA) to approximate complement-fixing antibody titers in serum from patients with coccidioidomycosis. Diagn. Microbiol. Infect. Dis. 2021, 99, 115198. [Google Scholar] [CrossRef]
  51. Zimmermann, C.R.; Johnson, S.M.; Martens, G.W.; White, A.G.; Pappagianis, D. Cloning and expression of the complement fixation antigen-chitinase of Coccidioides immitis. Infect. Immun. 1996, 64, 4967–4975. [Google Scholar] [CrossRef] [Green Version]
  52. Mead, H.L.; Roe, C.C.; Keppler, E.A.H.; Van Dyke, M.C.C.; Laux, K.L.; Funke, A.; Miller, K.J.; Bean, H.D.; Sahl, J.W.; Barker, B.M. Defining Critical Genes During Spherule Remodeling and Endospore Development in the Fungal Pathogen, Coccidioides posadasii. Front. Genet. 2020, 11, 483. [Google Scholar] [CrossRef] [PubMed]
  53. Bean, H. Identifying volatile breathmarkers for a Valley fever breath test. In Proceedings of the Coccidioidomycosis Study Group Annual Meeting, Davis, CA, USA, 5 April 2019. [Google Scholar]
  54. Jasbi, P.; Mitchell, N.M.; Shi, X.; Grys, T.E.; Wei, Y.; Liu, L.; Lake, D.F.; Gu, H. Coccidioidomycosis Detection Using Targeted Plasma and Urine Metabolic Profiling. J. Proteome Res. 2019, 18, 2791–2802. [Google Scholar] [CrossRef] [PubMed]
  55. Smith, C.E.; Beard, R.R. Varieties of Coccidioidal Infection in Relation to the Epidemiology and Control of the Diseases. Am. J. Public Health Nations Health 1946, 36, 1394–1402. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Bays, D.J.; Thompson, G.R.; Reef, S.; Snyder, L.; Freifeld, A.J.; Huppert, M.; Salkin, D.; Wilson, M.D.; Galgiani, J.N. Natural History of Disseminated Coccidioidomycosis: Examination of the Veterans Affairs–Armed Forces Database. Clin. Infect. Dis. 2020. [Google Scholar] [CrossRef] [PubMed]
  57. Ampel, N.M.; Giblin, A.; Mourani, J.P.; Galgiani, J.N. Factors and Outcomes Associated with the Decision to Treat Primary Pulmonary Coccidioidomycosis. Clin. Infect. Dis. 2009, 48, 172–178. [Google Scholar] [CrossRef] [PubMed]
  58. Blair, J.E.; Chang, Y.-H.H.; Cheng, M.-R.; Vaszar, L.T.; Vikram, H.R.; Orenstein, R.; Kusne, S.; Ho, S.; Seville, M.T.; Parish, J.M. Characteristics of Patients with Mild to Moderate Primary Pulmonary Coccidioidomycosis. Emerg. Infect. Dis. 2014, 20, 983–990. [Google Scholar] [CrossRef] [Green Version]
  59. Graybill, J.R.; Stevens, D.A.; Galgiani, J.N.; Dismukes, W.E.; Cloud, G.A. Itraconazole treatment of coccidioidomycosis: NIAID Mycoses Study Group. Am. J. Med. 1990, 89, 282–290. [Google Scholar] [CrossRef]
  60. Catanzaro, A.; Galgiani, J.N.; Levine, B.E.; Sharkey-Mathis, P.K.; Fierer, J.; Stevens, D.A.; Chapman, S.W.; Cloud, G. Fluconazole in the treatment of chronic pulmonary and nonmeningeal disseminated coccidioidomycosis: NIAID Mycoses Study Group. Am. J. Med. 1995, 98, 249–256. [Google Scholar] [CrossRef]
  61. Galgiani, J.N.; Catanzaro, A.; Cloud, G.A.; Johnson, R.H.; Williams, P.L.; Mirels, L.F.; Nassar, F.; Lutz, J.E.; Stevens, D.A.; Sharkey, P.K.; et al. Comparison of Oral Fluconazole and Itraconazole for Progressive, Nonmeningeal Coccidioidomycosis. Ann. Intern. Med. 2000, 133, 676–686. [Google Scholar] [CrossRef]
  62. Kim, M.M.; Vikram, H.R.; Kusne, S.; Seville, M.T.; Blair, J. Treatment of Refractory Coccidioidomycosis With Voriconazole or Posaconazole. Clin. Infect. Dis. 2011, 53, 1060–1066. [Google Scholar] [CrossRef] [Green Version]
  63. Ramani, R.; Chaturvedi, V. Antifungal susceptibility profiles of Coccidioides immitis and Coccidioides posadasii from endemic and non-endemic areas. Mycopathologia 2007, 163, 315–319. [Google Scholar] [CrossRef] [PubMed]
  64. Thompson, G.R.; Barker, B.M.; Wiederhold, N.P. Large-Scale Evaluation of In Vitro Amphotericin B, Triazole, and Echinocandin Activity against Coccidioides Species from U.S. Institutions. Antimicrob. Agents Chemother. 2017, 61, e02634-16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Thompson, G.R., 3rd; Lewis, J.S., 2nd; Nix, D.E.; Patterson, T.F. Current Concepts and Future Directions in the Pharmacology and Treatment of Coccidioidomycosis. Med. Mycol. 2019, 57 (Suppl. 1), S76–S84. [Google Scholar] [CrossRef]
  66. Strushkevich, N.; Usanov, S.A.; Park, H.-W. Structural Basis of Human CYP51 Inhibition by Antifungal Azoles. J. Mol. Biol. 2010, 397, 1067–1078. [Google Scholar] [CrossRef] [PubMed]
  67. Andes, D.R.; Azie, N.; Yang, H.; Harrington, R.; Kelley, C.; Tan, R.-D.; Wu, E.Q.; Franks, B.; Kristy, R.; Lee, E.; et al. Drug-Drug Interaction Associated with Mold-Active Triazoles among Hospitalized Patients. Antimicrob. Agents Chemother. 2016, 60, 3398–3406. [Google Scholar] [CrossRef] [Green Version]
  68. Davis, M.R.; Nguyen, M.-V.H.; Donnelley, M.; Iii, G.R.T. Tolerability of long-term fluconazole therapy. J. Antimicrob. Chemother. 2018, 74, 768–771. [Google Scholar] [CrossRef]
  69. Bercovitch, R.S.; Catanzaro, A.; Schwartz, B.S.; Pappagianis, D.; Watts, D.; Ampel, N.M. Coccidioidomycosis During Pregnancy: A Review and Recommendations for Management. Clin. Infect. Dis. 2011, 53, 363–368. [Google Scholar] [CrossRef] [Green Version]
  70. Thompson, G.R., 3rd; Bays, D.; Cohen, S.H.; Pappagianis, D. Fluoride excess in coccidioidomycosis patients receiving long-term antifungal therapy: An assessment of currently available triazoles. Antimicrob. Agents Chemother. 2012, 56, 563–564. [Google Scholar] [CrossRef] [Green Version]
  71. Hedrick, J.; Droz, N. Voriconazole-Induced Periostitis. N. Engl. J. Med. 2019, 381, e30. [Google Scholar] [CrossRef]
  72. Boughton, C.; Taylor, D.; Ghataore, L.; Taylor, N.; Whitelaw, B.C. Mineralocorticoid hypertension and hypokalaemia induced by posaconazole. Endocrinol. Diabetes Metab. Case Rep. 2018, 2018. [Google Scholar] [CrossRef]
  73. Thompson, G.R., 3rd; Beck, K.R.; Patt, M.; Kratschmar, D.V.; Odermatt, A. Posaconazole-Induced Hypertension Due to Inhibition of 11beta-Hydroxylase and 11beta-Hydroxysteroid Dehydrogenase 2. J. Endocr. Soc. 2019, 3, 1361–1366. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Rauseo, A.M.; Coler-Reilly, A.; Larson, L.; Spec, A. Hope on the Horizon: Novel Fungal Treatments in Development. Open Forum Infect. Dis. 2020, 7, ofaa016. [Google Scholar] [CrossRef] [Green Version]
  75. Hector, R.F.; Zimmer, B.L.; Pappagianis, D. Evaluation of nikkomycins X and Z in murine models of coccidioidomycosis, histoplasmosis, and blastomycosis. Antimicrob. Agents Chemother. 1990, 34, 587–593. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Li, R.K.; Rinaldi, M.G. In Vitro Antifungal Activity of Nikkomycin Z in Combination with Fluconazole or Itraconazole. Antimicrob. Agents Chemother. 1999, 43, 1401–1405. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Nix, D.E.; Swezey, R.R.; Hector, R.; Galgiani, J.N. Pharmacokinetics of Nikkomycin Z after Single Rising Oral Doses. Antimicrob. Agents Chemother. 2009, 53, 2517–2521. [Google Scholar] [CrossRef] [Green Version]
  78. Shubitz, L.F.; Roy, M.E.; Nix, D.E.; Galgiani, J.N. Efficacy of Nikkomycin Z for respiratory coccidioidomycosis in naturally infected dogs. Med. Mycol. 2013, 51, 747–754. [Google Scholar] [CrossRef] [Green Version]
  79. Shubitz, L.F.; Trinh, H.T.; Perrill, R.H.; Thompson, C.M.; Hanan, N.J.; Galgiani, J.N.; Nix, D.E. Modeling Nikkomycin Z Dosing and Pharmacology in Murine Pulmonary Coccidioidomycosis Preparatory to Phase 2 Clinical Trials. J. Infect. Dis. 2014, 209, 1949–1954. [Google Scholar] [CrossRef] [Green Version]
  80. 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. 2017, 73, 404–408. [Google Scholar] [CrossRef] [Green Version]
  81. Wiederhold, N.P.; Shubitz, L.F.; Najvar, L.K.; Jaramillo, R.; Olivo, M.; Catano, G.; Trinh, H.T.; Yates, C.M.; Schotzinger, R.J.; Garvey, E.P.; et al. The Novel Fungal Cyp51 Inhibitor VT-1598 Is Efficacious in Experimental Models of Central Nervous System Coccidioidomycosis Caused by Coccidioides posadasii and Coccidioides immitis. Antimicrob. Agents Chemother. 2018, 62, 02258–17. [Google Scholar] [CrossRef] [Green Version]
  82. Oliver, J.D.; Sibley, G.E.M.; Beckmann, N.; Dobb, K.S.; Slater, M.J.; McEntee, L.; Du Pré, S.; Livermore, J.; Bromley, M.J.; Wiederhold, N.P.; et al. F901318 represents a novel class of antifungal drug that inhibits dihydroorotate dehydrogenase. Proc. Natl. Acad. Sci. USA 2016, 113, 12809–12814. [Google Scholar] [CrossRef] [Green Version]
  83. Wiederhold, N.P. Review of the Novel Investigational Antifungal Olorofim. J. Fungi 2020, 6, 122. [Google Scholar] [CrossRef] [PubMed]
  84. Wiederhold, N.P.; Najvar, L.K.; Jaramillo, R.; Olivo, M.; Birch, M.; Law, D.; Rex, J.H.; Catano, G.; Patterson, T.F. The Orotomide Olorofim Is Efficacious in an Experimental Model of Central Nervous System Coccidioidomycosis. Antimicrob. Agents Chemother. 2018, 62. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Kirkland, T.N. The Quest for a Vaccine Against Coccidioidomycosis: A Neglected Disease of the Americas. J. Fungi 2016, 2, 34. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Barnato, A.E.; Sanders, G.D.; Owens, D.K. Cost-effectiveness of a potential vaccine for Coccidioides immitis. Emerg. Infect. Dis. 2001, 7, 797–806. [Google Scholar] [CrossRef] [PubMed]
  87. Pappagianis, D. Evaluation of the protective efficacy of the killed Coccidioides immitis spherule vaccine in human: The Valley Fever Vaccine Study Group. Am. Rev. Respir. Dis. 1993, 148, 656–660. [Google Scholar] [CrossRef] [PubMed]
  88. Da Silva, L.B.R.; Taborda, C.P.; Nosanchuk, J.D. Advances in Fungal Peptide Vaccines. J. Fungi 2020, 6, 119. [Google Scholar] [CrossRef]
  89. Narra, H.P.; Shubitz, L.F.; Mandel, M.A.; Trinh, H.T.; Griffin, K.; Buntzman, A.S.; Frelinger, J.A.; Galgiani, J.N.; Orbach, M.J. A Coccidioides posadasii CPS1 Deletion Mutant Is Avirulent and Protects Mice from Lethal Infection. Infect. Immun. 2016, 84, 3007–3016. [Google Scholar] [CrossRef] [Green Version]
  90. Hung, C.-Y.; Castro-Lopez, N.; Cole, G.T. Vaccinated C57BL/6 Mice Develop Protective and Memory T Cell Responses to Coccidioides posadasii Infection in the Absence of Interleukin-10. Infect. Immun. 2013, 82, 903–913. [Google Scholar] [CrossRef] [Green Version]
  91. Hung, C.Y.; Zhang, H.; Castro-Lopez, N.; Ostroff, G.R.; Khoshlenar, P.; Abraham, A.; Cole, G.T.; Negron, A.; Forsthuber, T.; Peng, T.; et al. Glucan-Chitin Particles Enhance Th17 Response and Improve Protective Efficacy of a Multivalent Antigen (rCpa1) against PulmonaryCoccidioides posadasiiInfection. Infect. Immun. 2018, 86, 00070-18. [Google Scholar] [CrossRef] [Green Version]
  92. Campuzano, A.; Zhang, H.; Ostroff, G.; Dias, L.S.; Wüthrich, M.; Klein, B.S.; Yu, J.; Lara, H.H.; Lopez-Ribot, J.L.; Hung, C.Y. CARD9-Associated Dectin-1 and Dectin-2 Are Required for Protective Immunity of a Multivalent Vaccine against Coccidioides posadasii Infection. J. Immunol. 2020, 204, 3296–3306. [Google Scholar] [CrossRef]
Table 1. Comparison of minimum inhibitory concentrations (µg/mL) of triazole antifungals.
Table 1. Comparison of minimum inhibitory concentrations (µg/mL) of triazole antifungals.
ParameterFluconazole
(n = 581)
Itraconazole
(n = 486)
Voriconazole
(n = 499)
Posaconazole
(n = 377)
MIC5080.2500.1250.125
MIC90160.5000.2500.250
GM MIC7.710.2450.1070.141
MIC50: minimum inhibitory concentration 50%. MIC90: minimum inhibitory concentration 90%. GM MIC: geometric mean minimum inhibitory concentration. Parentheses contain number of isolates tested. Adapted from Thompson et al. [62].
Table 2. Antifungals on the horizon.
Table 2. Antifungals on the horizon.
AgentMechanism of ActionHalf-LifeRoute of Delivery
Nikkomycin Zcompetitive inhibition of chitin synthase1–2 hIV and oral
VT-1598inhibition of 14-α-demethylase (CYP51)24 horal
Olorofimreversible inhibition pyrimidine biosynthesis20–30 hIV (with vehicle) and oral
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Ampel, N.M. Coccidioidomycosis: Changing Concepts and Knowledge Gaps. J. Fungi 2020, 6, 354. https://doi.org/10.3390/jof6040354

AMA Style

Ampel NM. Coccidioidomycosis: Changing Concepts and Knowledge Gaps. Journal of Fungi. 2020; 6(4):354. https://doi.org/10.3390/jof6040354

Chicago/Turabian Style

Ampel, Neil M. 2020. "Coccidioidomycosis: Changing Concepts and Knowledge Gaps" Journal of Fungi 6, no. 4: 354. https://doi.org/10.3390/jof6040354

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