Abstract
Invasive fungal infections are responsible for a significant disease burden worldwide. Drugs to treat these infections are limited to only four unique classes, and despite these available treatments, mortality rates remain unacceptably high. In this review, we will discuss antifungal drug screening and how the approach to identifying novel compounds needs move away from traditional growth-based assays in order to meet the demand for new drugs. We highlight specific examples of creative screening strategies that increase the likelihood of identifying compounds with desired activities and provide perspective to inspire development of novel screens for the identification of first-in-class antifungals.
Introduction
Invasive fungal infections (IFIs) are a significant cause of morbidity and mortality worldwide, leading to an estimated one and a half million deaths per year [1]. In addition to the importance of invasive infections, the role of fungi and the mycobiome in the development of other diseases is becoming an area of interest [2]. Currently, there are four classes of antifungal drugs used clinically; of these, only three of which are effective as monotherapies [3]. The development of antifungal drugs has been slow compared that of other types of drugs. For example, from 2000 to 2015, 18 first-in-class drugs were approved for the use of solid tumor cancers [4]. In contrast, only a single novel class of antifungal drugs, the echinocandins, were introduced during this time; specifically, caspofungin was approved for invasive aspergillosis in 2001 [5]. The slow pace of antifungal drug development is due in part to a variety of factors: a shrinking interest of big pharma in antimicrobials [6,7]; the conservation of many biological pathways between human and fungi; and the difficulty and expense of doing properly powered clinical trials [3]. High mortality rates of IFIs, toxicity of available antifungal, and intrinsic and emergent drug resistance highlight the urgent need for new antifungal drugs.
The ‘golden age’ of antibacterial discovery has been followed by a fallow stretch characterized by low-yield screening efforts. An important reason for this lull in discovery is that many of the ‘low hanging fruit’-type compounds have been identified. As a result, application of the same growth assay-based screening strategies has led to the repeated rediscovery of the same classes compounds [8,9]. Antifungal drug development is coming to the same fate, with the same compound families and targets being identified repeatedly. Screening experiments have essentially two variables: library content and screening assay readout. Either of these variables could contribute to the ‘discovery bottle-neck’. Here we propose that moving beyond simple cell density/growth-based assays may improve our ability to identify new chemical matter within old chemical libraries.
Growth-based assays
The use of culture optical density (OD) as a readout of fungal cell growth is quick, cheap, and convenient for screening compounds against yeast and has been frequently used in drug discovery. However, these assays can be less sensitive than alternative methods and are not amenable to screening organisms with filamentous growth. Molds are a particular challenge for high throughput screening. Hyphal cultures are heterogenous, cannot be inoculated after germination, and present a high risk for contamination of equipment and facilities. OD measurements of filamentous cultures can be unreliable for two reasons. First, cells are not homogenously distributed within the well. Second, these cultures frequently form biofilms on the surface of the liquid. Because of these technical issues, traditional OD assays can only identify compounds that completely inhibit germination or growth of filamentous fungi and, thus, have poor sensitivity.
Alternative measures of cell growth can provide a more robust measurement of inhibition of filamentous cultures. The blue resazurin molecule is metabolically reduced to the pink, fluorescent resorufin and has been used to screen Aspergillus fumigatus [10] as well as the biofilm stage of Candida albicans [11–13]. Similarly, metabolism of the tetrazolium salt XTT has been used in screens for compounds active against C. albicans biofilms [14]. Quantification of total ATP in a sample has also been used withC. albicans biofilms [15,16], A. fumigatus, and the dematiaceous mold Exserohilum rostratum [17]. While these alternatives to OD provide methods to detect growth inhibition in a wider range of organisms or biological states, it is important to consider that the readouts can be altered by changes in metabolism that don’t necessarily reflect growth inhibition (Table 1). Ultimately, the desired effect of an antifungal is the inhibition of growth; however, measuring other cellular responses or coupling these growth assays with other approaches can provide more sensitive detection of molecules with antifungal activity.
Table 1.
Strengths and limitations of screening assays
| Assay | Strengths | Limitations |
|---|---|---|
| Optical density |
|
|
| Metabolic reporters |
|
|
| Reporter strains |
|
|
| Adenylate kinase assay |
|
|
| In vivo screening |
|
|
Assays designed to target specific pathways in fungal cells
As a result of decades of research on the biology of pathogenic fungi, a wealth of knowledge about pathways required for growth and virulence is available. Designing assays to specifically detect molecules that interfere with these pathways is an effective approach to identifying mechanistic novel molecules, particularly if these assays are tailored to whole cell screening. For example, C. albicans encodes 115 glycosylphosphatidylinositol (GPI)-anchored proteins that are both critical to cell wall integrity and adhesion to host cells [18]. As such, many of the genes involved in GPI protein biosynthesis are essential and, thus, attractive as an antifungal drug target. To identify a first-in-class lead compound that inhibits GPI biosynthesis, a Saccharomyces cerevisiae reporter strain expressing a fusion protein of bacterial cephalosporinase to the GPI-anchored cell wall protein, Cwp2, was engineered [19]. Compounds were screened for those that reduced cephalosporinase activity in the cell wall fraction. This screen identified 1-[4-butylbenzyl]isoquinoline (BIQ) which was then optimized to the current clinical candidate APX001A. APX001A is a potent, broad spectrum antifungal with activity against yeast and mold, including difficult to treat organisms such as Scedosporium, Rhizopus [20,21], and the emerging drug resistant pathogen, Candida auris [22,23]. This compound is currently in phase 2 clinical trials for invasive candidiasis including C. auris (NCT03604705; NCT04148287). Notably, this screening methodology detected a 50% decrease in cephalosporinase activity at an 8-fold lower concentration than that which inhibits growth. This exemplifies how non-growth-based assays can identify novel compounds that would be missed in growth-based screens. The use of other reporter strains has been applied to identify molecules that interfere with critical pathways such as zinc and iron homeostasis [24,25] and the sterol regulatory transcription factor Upc2 [26]. These screening strategies bias compound identification to those that act on specific pathways and, thereby, reduce the likelihood of the ‘rediscovery’ phenomenon.
Distinguishing between fungicidal and fungistatic molecules
IFIs largely affect patients with immune deficiencies[1].This places a large dependence on drug treatment to clear infections without the assistance of immune cells available in normal hosts. Thus, fungicidal drugs which kill cells, rather than inhibit growth, are ideal for antifungals. In support of this, clinical studies of cryptococcal meningitis show that early clearance of organism from the cerebral spinal fluid using fungicidal drugs is associated with better patient outcomes [27–29]. Traditional growth-based assays do not distinguish between fungicidal and fungistatic molecules. To address this limitation in specificity, the adenylate kinase (AK) assay has been used to specifically identify fungicidal molecules in the high throughput settings. The AK assay measures the release of the ubiquitous cytosolic enzyme, AK, as a readout of cell lysis [30]. The AK assay is extremely sensitive and can detect cell lysis at eightfold lower concentrations of fungi-lytic drugs than traditional growth assays using OD [30]. Furthermore, the detection of a positive signal increases the linear range of the assay versus most growth assays which detect a reduction in signal. This sensitivity allows for detection of fungi-lytic molecules that would be missed by other growth assays; however, it will not detect compounds such as azoles, which do not induce cell lysis [30]. The AK assay identified the estrogen receptor antagonists, tamoxifen, and related compounds as promising antifungals [30]. While tamoxifen had previously identified antifungal activity against S. cerevisiae [31] and C. albicans [32], the identification of this drug in a screen to validate the AK assay reinvigorated interest in this drug [30]. Recently tamoxifen entered phase II clinical trial as an adjunct therapy with amphotericin B and fluconazole for cryptococcal meningitis (NCT03112031). Coupling the AK assay with secondary screens to identify cell wall active molecules led to the identification of a novel class of anti-cryptococcal molecules that block signaling through the cell wall integrity pathway [33,34].
Screening under physiologically relevant conditions
The use of whole animals in drug screening offers the unique ability to counter-screen for toxicity while screening in an actual infection setting. Antifungal screens have been implemented using the nematode, Caenorhabditis elegans [35,36]. In this model, worms are co-inoculated into plates with C. albicans, then wells are imaged with an automated imaging system, and the viability of the worms is assessed manually [36]. Notably, this screening technique can identify compounds that inhibit in vitro but not in vivo growth of C. albicans, potentially saving a lot of time following up on molecules that will fail in preclinical models of disease. The larval zebra fish, Danio rerio, has been well developed as a model system for many human fungal pathogens (reviewed in Ref. [37]). Although high throughput screens have not been performed for anti-infectives, large-scale compound screens have been performed on mutant fish to find therapeutics for specific genetic deficiencies [38,39]. Fungal infection models require a microinjection inoculation technique which is more laborious than co-culture; however, automated methods provide more potential for larger, more rapid screens [40].
While whole animal screens are not always feasible and present their own limitations (Table 1), in vitro screens can be designed to better represent in vivo conditions. Designing screens that target specific metabolic states is an example of emulating in vivo conditions in vitro. Using lactate as the sole carbon source can identify inhibitors of isocitrate lyase, an enzyme required for assimilation of C2 carbon sources and critical for pathogenesis [41,42]. Metabolic manipulations with the growth conditions have been successfully used to identify several antibiotics [43,44] and has the same potential for fungi if applied to larger scale screens. Another important aspect of an infection is the gas environment. The ability of Cryptococcus neoformans to adapt to physiological carbon dioxide correlates with the ability to cause disease in mice [45]. Similarly, low oxygen is an important characteristic of pulmonary aspergillosis [46], and the ability to adapt to low oxygen is correlated with disease severity in murine models of aspergillosis [47]. These conditions can be explored in drug screens to better represent the environment encountered at the site of infection and could identify compounds that would be missed in screens under standard lab growth conditions.
Repurposing FDA approved compounds
The screening of FDA-approved drug libraries for drug repurposing has become an attractive approach to drug development. These drugs have already passed initial safety screening and data on in vivo pharmacokinetics and pharmacodynamics are already available [48]. Repurposing screens have been performed against many major fungal pathogens, including C. albicans [49–54], C. neoformans [49,55], A. fumigatus [50,56], and the emerging multidrug resistant C. auris [14,50,57,58]. A screen against Aspergillus nidulans using the Johns Hopkins Clinical Compounds library identified the antidepressant, Sertraline, as having modest anti-Aspergillus activity [56]. Follow-up studies established more potent activity against C. neoformans [59] and a clinical trial was performed for cryptococcal meningitis with sertraline as an adjunctive therapy to Amphotericin B and fluconazole [60]. Although this trial failed to show a survival benefit with adjunctive sertraline, bringing an antifungal to clinical trial is a success that should not be overlooked. This example, and tamoxifen described above, highlight how repurposing can expedite the development of antifungals. However repurposing drug libraries are generally small and have been screened many times over, so choosing a library carefully can avoid wasting resources on screening the same set of compounds over and over again.
Combination screens and antifungal resistance
Screening for drugs which enhance the activity of current antifungals is another approach to improving antifungal therapy. Combination therapy offers the possibility of using lower doses, thus lower toxicity, of current antifungals, providing more broad spectrum coverage and shifting fungistatic activity to fungicidal activity [61]. Robbins et al. [62] screened six antifungal drugs at sub-inhibitory concentrations, in combination with ~3600 small molecules, against four fungal species. This study identified several small molecules, including FDA-approved drugs that increase the efficacy of antifungals, and even potentiate the activity of antifungals in resistant strains [62]. Performing combination screens can also be done using a resistant strain directly, to identify compounds that can be used to treat resistant organisms and to reduce the identification of cross-resistant molecules. A screen using echinocandin resistant C. albicans identified the metal chelator DTPA as a potentiator of caspofungin activity [63] and treatment with DPTA and caspofungin together significantly increased survival of mice inoculated with echinocandin resistant C. albicans compared to each drug alone.
Future perspectives
Increasing the diversity of antifungal screening assays will allow investigators to cover more chemical space and accordingly improve the chances that novel antifungal scaffolds will be discovered. While the idea of an ‘ideal’ broad-spectrum antifungal is attractive, the search for the perfect can hinder the identification of the good. Although growth-based screens are simple to perform, we must consider the old adage ‘you get what you screen for’. Repeating the same assay will yield the same results. To reuse the famous definition of insanity widely mis-attributed to Albert Einstein, “the definition of insanity is repeating the same thing again and expecting a different result”. It is time that antifungal drug discovery diversifies its approaches and methods.
Acknowledgement
This work was supported by N.I.H. grants 1F32AI145160-01A1 (SRB) and 1R01AI120958 (DJK).
Footnotes
Conflict of interest
The authors have no competing interests regarding the material discussed in this review.
References
- 1.Brown GD, Denning DW, Gow NAR, Levitz SM, Netea MG, White TC: Hidden killers: human fungal infections. Sci Transl Med 2012, 4:165rv13. [DOI] [PubMed] [Google Scholar]
- 2.Aykut B, Pushalkar S, Chen R, Li Q, Abengozar R, Kim JI, Shadaloey SA, Wu D, Preiss P, Verma N et al. : The fungal mycobiome promotes pancreatic oncogenesis via activation of MBL. Nature 2019, 574:264–267. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Perfect JR: The antifungal pipeline: a reality check. Nat Rev Drug Discov 2017, 16:603–616. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Vivot A, Jacot J, Zeitoun J-D, Ravaud P, Crequit P, Porcher R: Clinical benefit, price and approval characteristics of FDA-approved new drugs for treating advanced solid cancer, 2000–2015. Ann Oncol 2017, 28:1111–1116. [DOI] [PubMed] [Google Scholar]
- 5.Balkovec JM, Hughes DL, Masurekar PS, Sable CA, Schwartz RE, Singh SB: Discovery and development of first in class antifungal caspofungin (CANCIDASJ)—a case study. Nat Prod Rep 2014, 31:15–34. [DOI] [PubMed] [Google Scholar]
- 6.Wanted: a reward for antibiotic development. Nat Biotechnol 2018, 36:555. [DOI] [PubMed] [Google Scholar]
- 7.Projan SJ: Why is big pharma getting out of antibacterial drug discovery? Curr Opin Microbiol 2003, 6:427–430. [DOI] [PubMed] [Google Scholar]
- 8.Silver LL: Challenges of antibacterial discovery. Clin Microbiol Rev 2011, 24:71–109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Brown ED, Wright GD: Antibacterial drug discovery in the resistance era. Nature 2016, 529:336–343. [DOI] [PubMed] [Google Scholar]
- 10.Monteiro MC, de la Cruz M, Cantizani J, Moreno C, Tormo JR, Mellado E, De Lucas JR, Asensio F, Valiante V, Brakhage AA et al. : A new approach to drug discovery. J Biomol Screen 2012, 17:542–549. [DOI] [PubMed] [Google Scholar]
- 11.Vila T, Lopez-Ribot JL: Screening the pathogen box for identification of Candida albicans biofilm inhibitors. Antimicrob Agents Chemother 2017, 61:1–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Delattin N, De Brucker K, Vandamme K, Meert E, Marchand A, Chaltin P, Cammue BPA, Thevissen K: Repurposing as a means to increase the activity of amphotericin B and caspofungin against Candida albicans biofilms. J Antimicrob Chemother 2014, 69:1035–1044. [DOI] [PubMed] [Google Scholar]
- 13.LaFleur MD, Lucumi E, Napper AD, Diamond SL, Lewis K: Novel high-throughput screen against Candida albicans identifies antifungal potentiators and agents effective against biofilms. J Antimicrob Chemother 2011, 66:820–826. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Wall Herrera, Lopez-Ribot: Repositionable compounds with antifungal activity against multidrug resistant Candida auris identified in the medicines for malaria venture’s pathogen box. J Fungi 2019, 5:92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Stylianou M, Uvell H, Lopes JP, Enquist P-A, Elofsson M, Urban CF: Novel high-throughput screening method for identification of fungal dimorphism blockers. J Biomol Screen 2015, 20:285–291. [DOI] [PubMed] [Google Scholar]
- 16.Watamoto T, Egusa H, Sawase T, Yatani H: Screening of pharmacologically active small molecule compounds identifies antifungal agents against candida biofilms. Front Microbiol 2015, 6:1–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Sun W, Park Y-D, Sugui JA, Fothergill A, Southall N, Shinn P, McKew JC, Kwon-Chung KJ, Zheng W, Williamson PR: Rapid identification of antifungal compounds against Exserohilum rostratum using high throughput drug repurposing screens. PLoS One 2013, 8:e70506. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Richard ML, Plaine A: Comprehensive analysis of glycosylphosphatidylinositol-anchored proteins in Candida albicans. Eukaryot Cell 2007, 6:119–133. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Tsukahara K, Hata K, Nakamoto K, Sagane K, Watanabe N, Kuromitsu J, Kai J, Tsuchiya M, Ohba F, Jigami Y et al. : Medicinal genetics approach towards identifying the molecular target of a novel inhibitor of fungal cell wall assembly. Mol Microbiol 2003, 48:1029–1042. [DOI] [PubMed] [Google Scholar]
- 20.Miyazaki M, Horii T, Hata K, Watanabe N, Nakamoto K, Tanaka K, Shirotori S, Murai N, Inoue S, Matsukura M et al. : In vitro activity of E1210, a novel antifungal, against clinically important yeasts and molds. Antimicrob Agents Chemother 2011, 55:4652–4658. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Hata K, Horii T, Miyazaki M, Watanabe N, Okubo M, Sonoda J, Nakamoto K, Tanaka K, Shirotori S, Murai N et al. : Efficacy of oral E1210, a new broad-spectrum antifungal with a novel mechanism of action, in murine models of candidiasis, aspergillosis, and fusariosis. Antimicrob Agents Chemother 2011, 55:4543–4551. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Berkow EL, Lockhart SR: Activity of novel antifungal compound APX001A against a large collection of Candida auris. J Antimicrob Chemother 2018, 73:3060–3062. [DOI] [PubMed] [Google Scholar]
- 23.Hager CL, Larkin EL, Long L, Zohra Abidi F, Shaw KJ, Ghannoum MA: In vitro and in vivo evaluation of the antifungal activity of APX001A/APX001 against Candida auris. Antimicrob Agents Chemother 2018, 62:e02319–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Simm C, Luan C-H, Weiss E, O’Halloran T: High-throughput screen for identifying small molecules that target fungal zinc homeostasis. PLoS One 2011, 6:e25136. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Simm C, May RC: Zinc and iron homeostasis: target-based drug screening as new route for antifungal drug development. Front Cell Infect Microbiol 2019, 9:1–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Gallo-Ebert C, Donigan M, Stroke IL, Swanson RN, Manners MT, Francisco J, Toner G, Gallagher D, Huang C, Gygax SE et al. : Novel antifungal drug discovery based on targeting pathways regulating the fungus-conserved Upc2 transcription factor. Antimicrob Agents Chemother 2014, 58:258–266. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Bicanic T, Muzoora C, Brouwer AE, Meintjes G, Longley N, Taseera K, Rebe K, Loyse A, Jarvis J, Bekker L et al. : Independent association between rate of clearance of infection and clinical outcome of HIV-associated cryptococcal meningitis: analysis of a combined cohort of 262 patients. Clin Infect Dis 2009, 49:702–709. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Day JN, Chau TTHH, Wolbers M, Mai PP, Dung NT, Mai NH, Phu NH, Nghia HD, Phong ND, Thai CQ et al. : Combination antifungal therapy for cryptococcal meningitis. N Engl J Med 2013, 368:1291–1302. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Larsen RA, Leal MA, Chan LS: Fluconazole compared with amphotericin B plus flucytosine for cryptococcal meningitis in AIDS. A randomized trial. Ann Intern Med 1990, 113:183–187. [DOI] [PubMed] [Google Scholar]
- 30.Krysan DJ, Didone L: A high-throughput screening assay for small molecules that disrupt yeast cell integrity. J Biomol Screen 2008, 13:657–664. [DOI] [PubMed] [Google Scholar]
- 31.Wiseman H, Cannon M, Arnstein HRV: Observation and significance of growth inhibition of Saccharomyces cerevisiae (A224A) by the antioestrogen drug tamoxifen. Biochem Soc Trans 1989, 17:1038–1039. [DOI] [PubMed] [Google Scholar]
- 32.Beggs WH: Anti-Candida activity of the anti-cancer drug tamoxifen. Res Commun Chem Pathol Pharmacol 1993, 80:125–128. [PubMed] [Google Scholar]
- 33.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, 6:502–510. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Hartland K, Pu J, Palmer M, Dandapani S, Moquist PN, Munoz B, DiDone L, Schreiber SL, Krysan DJ: High-throughput screen in Cryptococcus neoformans identifies a novel molecular scaffold that inhibits cell wall integrity pathway signaling. ACS Infect Dis 2016, 2:93–102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Okoli I, Coleman JJ, Tempakakis E, An WF, Holson E, Wagner F, Conery AL, Larkins-Ford J, Wu G, Stern A et al. : Identification of antifungal compounds active against Candida albicans using an improved high-throughput Caenorhabditis elegans assay. PLoS One 2009, 4:e7025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Breger J, Fuchs BB, Aperis G, Moy TI, Ausubel FM, Mylonakis E: Antifungal chemical compounds identified using a C. elegans pathogenicity assay. PLoS Pathog 2007, 3:e18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Rosowski E, Knox B, Archambault L, Huttenlocher A, Keller N, Wheeler R, Davis J: The zebrafish as a model host for invasive fungal infections. J Fungi 2018, 4:136. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Stern HM, Murphey RD, Shepard JL, Amatruda JF, Straub CT, Pfaff KL, Weber G, Tallarico JA, King RW, Zon LI: Small molecules that delay S phase suppress a zebrafish bmyb mutant. Nat Chem Biol 2005, 1:366–370. [DOI] [PubMed] [Google Scholar]
- 39.Peterson RT, Shaw SY, Peterson TA, Milan DJ, Zhong TP, Schreiber SL, MacRae CA, Fishman MC: Chemical suppression of a genetic mutation in a zebrafish model of aortic coarctation. Nat Biotechnol 2004, 22:595–599. [DOI] [PubMed] [Google Scholar]
- 40.Spaink HP, Cui C, Wiweger MI, Jansen HJ, Veneman WJ, Marín-Juez R, de Sonneville J, Ordas A, Torraca V, van der Ent W et al. : Robotic injection of zebrafish embryos for high-throughput screening in disease models. Methods 2013, 62:246–254. [DOI] [PubMed] [Google Scholar]
- 41.Cheah H-L, Lim V, Sandai D: Inhibitors of the glyoxylate cycle enzyme ICL1 in Candida albicans for potential use as antifungal agents. PLoS One 2014, 9:e95951. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Dunn MF, Ramirez-Trujillo JA, Hernandez-Lucas I: Major roles of isocitrate lyase and malate synthase in bacterial and fungal pathogenesis. Microbiology 2009, 155:3166–3175. [DOI] [PubMed] [Google Scholar]
- 43.Fahnoe KC, Flanagan ME, Gibson G, Shanmugasundaram V, Che Y, Tomaras AP: Non-traditional antibacterial screening approaches for the identification of novel inhibitors of the glyoxylate shunt in gram-negative pathogens. PLoS One 2012, 7:e51732. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Zlitni S, Ferruccio LF, Brown ED: Metabolic suppression identifies new antibacterial inhibitors under nutrient limitation. Nat Chem Biol 2013, 9:796–804. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Krysan DJ, Zhai B, Beattie SR, Misel KM, Wellington M, Lin X: Host carbon dioxide concentration is an independent stress for Cryptococcus neoformans that affects virulence and antifungal susceptibility. mBio 2019, 10:1–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Grahl N, Puttikamonkul S, Macdonald JM, Gamcsik MP, Ngo LY, Hohl TM, Cramer RA: In vivo hypoxia and a fungal alcohol dehydrogenase influence the pathogenesis of invasive pulmonary aspergillosis. PLoS Pathog 2011, 7:e1002145. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Kowalski CH, Beattie SR, Fuller KK, McGurk EA, Tang Y-W, Hohl TM, Obar JJ, Cramer RA: Heterogeneity among isolates reveals that fitness in low oxygen correlates with Aspergillus fumigatus virulence. mBio 2016, 7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Ashburn TT, Thor KB: Drug repositioning: identifying and developing new uses for existing drugs. Nat Rev Drug Discov 2004, 3:673–683. [DOI] [PubMed] [Google Scholar]
- 49.Spitzer M, Griffiths E, Blakely KM, Wildenhain J, Ejim L, Rossi L, De Pascale G, Curak J, Brown E, Tyers M et al. : Cross-species discovery of syncretic drug combinations that potentiate the antifungal fluconazole. Mol Syst Biol 2011, 7:499. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Mamouei Z, Alqarihi A, Singh S, Xu S, Mansour MK, Ibrahim AS, Uppuluri P: Alexidine dihydrochloride has broad-spectrum activities against diverse fungal pathogens. mSphere 2018, 3:1–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Kim K, Zilbermintz L, Martchenko M: Repurposing FDA approved drugs against the human fungal pathogen, Candida albicans. Ann Clin Microbiol Antimicrob 2015, 14:32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Holmes AR, Keniya MV, Ivnitski-Steele I, Monk BC, Lamping E, Sklar LA, Cannon RD: The monoamine oxidase A inhibitor clorgyline is a broad-spectrum inhibitor of fungal ABC and MFS transporter efflux pump activities which reverses the azole resistance of Candida albicans and Candida glabrata clinical isolates. Antimicrob Agents Chemother 2012, 56:1508–1515. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Bernardo SM, Allen CP, Waller A, Young SM, Oprea T, Sklar LA, Lee SA: An automated high-throughput cell-based multiplexed flow cytometry assay to identify novel compounds to target Candida albicans virulence-related proteins. PLoS One 2014, 9:e110354. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Siles SA, Srinivasan A, Pierce CG, Lopez-Ribot JL, Ramasubramanian AK: High-throughput screening of a collection of known pharmacologically active small compounds for identification of Candida albicans biofilm inhibitors. Antimicrob Agents Chemother 2013, 57:3681–3687. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Butts A, DiDone L, Koselny K, Baxter BK, Chabrier-Rosello Y, Wellington M, Krysan DJ: A repurposing approach identifies off-patent drugs with fungicidal cryptococcal activity, a common structural chemotype, and pharmacological properties relevant to the treatment of cryptococcosis. Eukaryot Cell 2013, 12:278–287. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Zhai B, Zhou H, Yang L, Zhang J, Jung K, Giam CZ, Xiang X, Lin X: Polymyxin B, in combination with fluconazole, exerts a potent fungicidal effect. J Antimicrob Chemother 2010, 65:931–938. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Wall G, Chaturvedi AK, Wormley FL, Wiederhold NP, Patterson HP, Patterson TF, Lopez-Ribot JL: Screening a repurposing library for inhibitors of multidrug-resistant Candida auris identifies ebselen as a repositionable candidate for antifungal drug development. Antimicrob Agents Chemother 2018, 62:1–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.de Oliveira HC, Monteiro MC, Rossi SA, Pemán J, Ruiz-Gaitán A, Mendes-Giannini MJS, Mellado E, Zaragoza O: Identification of off-patent compounds that present antifungal activity against the emerging fungal pathogen Candida auris. Front Cell Infect Microbiol 2019, 9:1–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Zhai B, Wu C, Wang L, Sachs MS, Lin X: The antidepressant sertraline provides a promising therapeutic option for neurotropic cryptococcal infections. Antimicrob Agents Chemother 2012, 56:3758–3766. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Rhein J, Huppler Hullsiek K, Tugume L, Nuwagira E, Mpoza E, Evans EE, Kiggundu R, Pastick KA, Ssebambulidde K, Akampurira A et al. : Adjunctive sertraline for HIV-associated cryptococcal meningitis: a randomised, placebo-controlled, double-blind phase 3 trial. Lancet Infect Dis 2019, 19:843–851. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Polak A: The past, present and future of antimycotic combination therapy. Mycoses 1999, 42:355–370. [DOI] [PubMed] [Google Scholar]
- 62.Robbins N, Spitzer M, Yu T, Cerone RP, Averette AK, Bahn Y-S, Heitman J, Sheppard DC, Tyers M, Wright GD: An antifungal combination matrix identifies a rich pool of adjuvant molecules that enhance drug activity against diverse fungal pathogens. Cell Rep 2015, 13:1481–1492. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Polvi EJ, Averette AF, Lee SC, Kim T, Bahn Y-S, Veri AO, Robbins N, Heitman J, Cowen LE: Metal chelation as a powerful strategy to probe cellular circuitry governing fungal drug resistance and morphogenesis. PLoS Genet 2016, 12: e1006350. [DOI] [PMC free article] [PubMed] [Google Scholar]
