244
Views
2
CrossRef citations to date
0
Altmetric
Review

Novel avenues for identification of new antifungal drugs and current challenges

Pages 949-968 | Received 02 Feb 2022, Accepted 30 Jun 2022, Published online: 21 Jul 2022

References

  • The Fungal BM. Kingdom: diverse and essential roles in earth’s ecosystem. Washington (DC): American Society for Microbiology; 2008 [cited 7 Jan 2022]. Available from: https://www.ncbi.nlm.nih.gov/books/NBK559443
  • Fowler S, Roush R, Wise J Diversity of microbes, fungi, and protists. In: concepts of biology, openstax CNX, 2019, chapter 13. cited 2022 Jan 07. Available from: https://opentextbc.ca/conceptsofbiologyopenstax/chapter/fungi/.
  • Prasad R. Fungal Nanotechnology: Applications in Agriculture, Industry, and Medicine. Cham, Switzerland: Springer International Publishing AG. 2017.
  • Singh J, and Gehlot P. New and future developments in microbial biotechnology and bioengineering. Amsterdam, Netherlands: Elsevier; 2021.
  • Ghosh S, Ahmad R, Zeyaullah M, et al. Microbial nano-factories: synthesis and biomedical applications. Front Chem. 2021;9:626834.
  • Fungal Infections. MSD and the MSD manuals. NJ: Merck and Co., Inc., Kenilworth; cited 07 Jan 2022 Available from: https://www.msdmanuals.com/home/infections/fungal-infections.
  • Bongomin F, Gago S, Oladele RO, Denning DW. Global and multi-national prevalence of fungal diseases-estimate precision. J Fungi. 2017;3(4):57.
  • Pristov KE, and Ghannoum MA. Resistance of Candida to azoles and echinocandins worldwide. Clin Microbiol Infect. 2019;25(7):792–798.
  • Frias-De-Leon MG, Pinto-Almazan R, Hernandez-Castro R, et al. Epidemiology of systemic mycoses in the COVID-19 pandemic. J Fungi. 2021;7(7):556.
  • Roudbary M, Kumar S, Kumar A, et al. Overview on the prevalence of fungal infections, immune response, and microbiome role in COVID-19 patients. J Fungi. 2021;7(9):720.
  • Sabino R, Verissimo C. Novel clinical and laboratorial challenges in aspergillosis. Microorganisms. 2022;10(2):259.
  • Fisher MC, Alastruey-Izquierdo A, and Berman J, et al. Tackling the emerging threat of antifungal resistance to human health. Nat Rev Microbiol. 2022 doi:10.1038/s41579-022-00720-1.
  • Global Antimicrobial Resistance Surveillance System (GLASS) early implementation protocol for the inclusion of Candida spp. Geneva: world health organization, 2019. [cited 07 Jan 2022]. Available from: https://www.who.int/publications/i/item/WHO-WSI-AMR-2019.4
  • Antifungal Expert WHO Group on identifying priority fungal pathogens: meeting report. Geneva: world health organization; 2020. [cited 07 Jan 2022]. Available from: https://www.who.int/publications/i/item/9789240006355
  • Global antimicrobial resistance and use surveillance system (GLASS) report 2021. Geneva: World Health Organization. [cited 07 Jan 2022]. Available from: https://www.who.int/publications/i/item/9789240027336
  • Li Y, Gao Y, Niu X, et al. A 5-year review of invasive fungal infection at an academic medical center. Front Cell Infect Microbiol. 2020;10:553648.
  • Firacative C. Invasive fungal disease in humans: are we aware of the real impact? Mem Inst Oswaldo Cruz. 2020;115:e200430.
  • Samaddar A, Sharma A. emergomycosis, an emerging systemic mycosis in immunocompromised patients: current trends and future prospects. Front Med. 2021;8:670731.
  • Kmeid J, Jabbour J-F, Kanj SS. Epidemiology and burden of invasive fungal infections in the countries of the Arab league. Journal of Infection and Public Health. 2020;13(12):2080–2086.
  • Donnelly JP, Chen SC, Kauffman CA, et al. revision and update of the consensus definitions of invasive fungal disease from the European organization for research and treatment of cancer and the mycoses study group education and research consortium. Clin Inf Dis. 2020;71(6):1367–1376.
  • Zhang H, Zhu A. Emerging invasive fungal infections: clinical features and controversies in diagnosis and treatment processes. Infect Drug Resist. 2020;13:607–615.
  • WHO: Mycotoxins. [cited 2022 Jan 08] Available from: https://www.who.int/news-room/fact-sheets/detail/mycotoxins
  • Ekwomadu T Toxic fungi: what’s in your food? Science today, 2016. [cited 08 Jan 2022]. Available from: https://sciencetoday.co.za/2016/11/14/toxic-fungi-whats-in-your-food
  • Eskola M, Kos G, Elliott CT, et al. Worldwide contamination of food-crops with mycotoxins: validity of the widely cited ‘FAO estimate’ of 25%. Crit Rev Food Sci Nutr. 2020;60(16):2773–2789.
  • Lelieveld H. Mycotoxins: a food safety crisis. food safety magazine e-digest 2015. [cited 08 Jan 2022]. Available from: https://www.foodsafetymagazine.com/enewsletter/mycotoxins-a-food-safety-crisis
  • Gruber-Dorninger C, Jenkins T, Schatzmayr G. Global mycotoxin occurrence in feed: a ten-year survey. Toxins (Basel). 2019;11(7):375.
  • Code List© FRAC 2022. [cited 13 June 2022]. Available from: https://www.frac.info/docs/default-source/publications/frac-code-list/frac-code-list-2022–final.pdf?sfvrsn=b6024e9a_2.
  • Jampilek J. Potential of agricultural fungicides for antifungal drug discovery. Expert Opin Drug Dis. 2016;11(1):1–9.
  • Griffith RK. Drugs used to treat fungal infections. In: Roche VF, Zito SW, Lemke T, et al., editors. Foye’s principles of medicinal chemistry, 8th edition. Philadelphia: Wolters Kluwer; 2019. p. 1260–1275.
  • Revankar SG Antifungal drugs. In: Merck manual professional version. [cited 10 Jan 2022]. Available from: https://www.merckmanuals.com/professional/infectious-diseases/fungi/antifungal-drugs
  • Chang YL, Yu SJ, Heitman J, et al. New facets of antifungal therapy. Virulence. 2017;8(2):222–236.
  • Jampilek J. Design and discovery of new antibacterial agents: advances, perspectives, challenges. Curr Med Chem. 2018;25(38):4972–5006.
  • Jampilek J, Kralova K. Application of nanotechnology in agriculture and food industry, its prospects and risks. Ecol Chem Eng S. 2015;22(3):321–361.
  • Jampilek J, Kralova K. Nano-antimicrobials: activity, benefits and weaknesses. In: Ficai A, Grumezescu AM, editors. Nanostructures for antimicrobial therapy. Amsterdam: Elsevier; 2017. p. 23–54.
  • Jampilek J, Kralova K. Impact of nanoparticles on toxigenic fungi. In: Rai M, Abd-Elsalam KA, editors. Nanomycotoxicology – treating mycotoxins in nano way. London: Academic Press & Elsevier; 2020. p. 309–348.
  • Jampilek J, Nanocomposites: KK. Synergistic nanotools for management mycotoxigenic fungi. In: Rai M, Abd-Elsalam KA, editors. Nanomycotoxicology – treating mycotoxins in nano way. London: Academic Press & Elsevier; 2020. p. 349–383.
  • Placha D, Jampilek J. graphenic materials for biomedical applications. Nanomaterials. 2019;9(12):1758.
  • Jampilek J, Kralova K. Advances in drug delivery nanosystems using graphene-based materials and carbon nanotubes. Materials. 2021;14(5):1059.
  • Ziental D, Mlynarczyk DT, Czarczynska-Goslinska B, et al. Photosensitizers mediated photodynamic inactivation against fungi. Nanomaterials. 2021;11(11):2883.
  • Jampilek J, Kralova K. Advances in nanostructures for antimicrobial therapy. Materials. 2022;15(7):2388.
  • Holmes AR, Cardno TS, Strouse JJ, et al. Targeting efflux pumps to overcome antifungal drug resistance. Future Med Chem. 2016;8(12):1485–1501.
  • Cornet M, Gaillardin C. pH signaling in human fungal pathogens: a new target for antifungal strategies. Eukaryot Cell. 2014;13(3):342–352.
  • Chatterjee S, Ghosh R, Mandal NC. Inhibition of biofilm- and hyphal- development, two virulent features of Candida albicans by secondary metabolites of an endophytic fungus Alternaria tenuissima having broad spectrum antifungal potential. Microbiol Res. 2020;232:126386.
  • Lohse MB, Gulati M, Craik CS, et al. Combination of antifungal drugs and protease inhibitors prevent Candida albicans biofilm formation and disrupt mature biofilms. Front Microbiol. 2020;11:1027.
  • Kathwate GH, Shinde RB, Mohan Karuppayil S. Non-antifungal drugs inhibit growth, morphogenesis and biofilm formation in Candida albicans. J Antibiot. 2021;74(5):346–353.
  • Ahmad Khan MS, Alshehrei F, Al-Ghamdi SB, et al., Virulence and biofilms as promising targets in developing antipathogenic drugs against candidiasis. Future Sci OA. 6(2): FSO440. 2020.
  • Jampilek J. Recent advances in design of potential quinoxaline anti-infectives. Curr Med Chem. 2014;21(38):4347–4373.
  • Pospisilova S, Kos J, and Michnova H, et al. Synthesis and spectrum of biological activities of novel N-arylcinnamamides. Int J Mol Sci. 2018;19(8):2318.
  • Talevi A. Multi-target pharmacology: possibilities and limitations of the “skeleton key approach” from a medicinal chemist perspective. Front Pharmacol. 2015;6:205.
  • Ramsay RR, Popovic-Nikolic MR, Nikolic K, et al. A perspective on multi-target drug discovery and design for complex diseases. Clin Transl Med. 2018;7:3.
  • Morphy JR. The challenges of multi-target lead optimization. designing multi-target drugs. Royal Society of Chemistry: London; 2012.
  • Yang T, Sui X, Yu B, et al. Recent advances in the rational drug design based on multi-target ligands. Curr Med Chem. 2020;27:4720–4740.
  • Pushpakom S, Iorio F, Eyers PA, et al. Drug repurposing: progress, challenges and recommendations. Nat Rev Drug Discov. 2019;18(1):41–58.
  • Rudrapal M, Khairnar SJ, Jadhav AG Drug repurposing (DR): an emerging approach in drug discovery. In: badria FA, editors. drug repurposing - hypothesis, molecular aspects and therapeutic applications, rijeka: intechopen, 2020. [cited 15 Jan 2022]. Available from: https://www.intechopen.com/chapters/72744.
  • Foletto VS, da Rosa TF, Serafin MB, et al., Repositioning of non-antibiotic drugs as an alternative to microbial resistance: a systematic review. Int J Antimicrob Agents. 58(3): 106380. 2021.
  • Schmidt RM, Rosenkranz HS. Antimicrobial activity of local anesthetics: lidocaine and procaine. J Inf Dis. 1970;121:597–607.
  • Pina-Vaz C, Rodrigues AG, and Sansonetty F, et al. Antifungal activity of local anesthetics against Candida species. Infect Dis Obstet Gynecol. 2000;8(3–4):124–137.
  • da C, Dos Santos O, and Cunha M, et al. In vitro synergistic activity of lidocaine and miconazole against Candida albicans. Acta Sci Health Sci. 2017;39(2):129–131.
  • Pospisilova S, Malik I, Bezouskova K, et al. Dibasic derivatives of phenylcarbamic acid as prospective antibacterial agents interacting with cytoplasmic membrane. Antibiotics. 2020;9(2):64.
  • Di Mambro T, Guerriero I, Aurisicchio L, et al. The yin and yang of current antifungal therapeutic strategies: how can we harness our natural defenses? Front Pharmacol. 2019;10:80.
  • Ulrich S, Ebel F. Monoclonal antibodies as tools to combat fungal infections. J Fungi. 2020;6(1):22.
  • Boniche C, Rossi SA, Kischkel B, et al., Immunotherapy against systemic fungal infections based on monoclonal antibodies. J Fungi. 6(1): 31. 2020.
  • Savarirajan D, Ramesh VM, and Muthaiyan A. In vitro antidermatophytic activity of bioactive compounds from selected medicinal plants. J Anal Sci Technol. 2021;12(1):53.
  • He W, Wu L, Gao Q, et al. Identification of AHBA biosynthetic genes related to geldanamycin biosynthesis in Streptomyces hygroscopicus 17997”. Curr Microbiol. 2006;52(3):197–203.
  • Cowen LE, Singh SD, Kohler JR, et al. Harnessing Hsp90 function as a powerful, broadly effective therapeutic strategy for fungal infectious disease. Proc Natl Acad Sci USA. 2009;106(8):2818–2823.
  • Whitesell L, Robbins N, Huang DS, et al., Structural basis for species-selective targeting of Hsp90 in a pathogenic fungus. Nat Commun. 10(1): 402. 2019.
  • Nakamura I, Yoshimura S, and Masaki T, et al. ASP2397: a novel antifungal agent produced by Acremonium persicinum MF-347833. J Antibiot. 2017;70:45–51.
  • Nakamura I, Ohsumi K, and Takeda S, et al. ASP2397 is a novel natural compound that exhibits rapid and potent fungicidal activity against Aspergillus species through a specific transporter. Antimicrob Agents Chemother. 2019;63(10):e02689–18.
  • Aguiar M, Orasch T, Misslinger M, et al., The siderophore transporters Sit1 and Sit2 are essential for utilization of ferrichrome-, ferrioxamine- and coprogen-type siderophores in Aspergillus fumigatus. J Fungi. 7(9): 768. 2021.
  • ClinicalTrials.gov. VL-2397 compared to standard first-line treatment for invasive aspergillosis (IA) in adults. [cited 2022 Jan 24 Available from: https://clinicaltrials.gov/ct2/show/NCT03327727.
  • Sari S, Kocak E, and Kart D, et al. Azole derivatives with naphthalene showing potent antifungal effects against planktonic and biofilm forms of Candida spp.: an in vitro and in silico study. Int Microbiol. 2021;24:93–102.
  • Oteseconazole, DrugBank. [cited 24 Jan 2022]. Available from: https://go.drugbank.com/drugs/DB13055
  • Pharmaceuticals M, 2020. [cited 24 Jan 2022]. Available from: https://www.fda.gov/media/141112/download
  • Wiederhold NP, Xu X, Wang A, et al. In vivo efficacy of VT-1129 against experimental Cryptococcal meningitis with the use of a loading dose-maintenance dose administration strategy. Antimicrob Agents Chemother. 2018;62(11):e01315–18.
  • Rezafungin, DrugBank. [cited 24 Jan 2022]. Available from: https://go.drugbank.com/drugs/DB16310
  • Ham YY, Lewis JS, Thompson GR. Rezafungin: a novel antifungal for the treatment of invasive candidiasis. Future Microbiol. 2021;16(1):27–36.
  • Thompson GR, Soriano A, Skoutelis A, et al. Rezafungin versus caspofungin in a phase 2, randomized, double-blind study for the treatment of candidemia and invasive Candidiasis: the STRIVE trial. Clin Infect Dis. 2021;73(11):e3647–55.
  • ClinicalTrials.gov. Study of rezafungin compared to standard antimicrobial regimen for prevention of invasive fungal diseases in adults undergoing allogeneic blood and marrow transplantation (ReSPECT). [cited 24 Jan 2022]. Available from: https://clinicaltrials.gov/ct2/show/NCT04368559.
  • Olorofim, DrugBank. [cited 24 Jan 2022]. Available from: https://go.drugbank.com/drugs/DB15245
  • ClinicalTrials.gov. Assessment of Varying oral dosing regimens for F901318 in healthy subjects. [cited 24 Jan 2022]. Available from: https://clinicaltrials.gov/ct2/show/NCT03340597.
  • Wiederhold NP. Review of the novel investigational antifungal olorofim. J Fungi. 2020;6(3):122.
  • Georgacopoulos O, Nunnally NS, and Ransom EM, et al. In vitro activity of novel antifungal olorofim against filamentous fungi and comparison to eight other antifungal agents. J Fungi. 2021;7(5):378.
  • Fosmanogepix, DrugBank. [cited 24 Jan 2022]. Available from: https://go.drugbank.com/drugs/DB15183
  • ClinicalTrials.gov. An efficacy and safety study of APX001 in non-neutropenic patients with Candidemia. [cited 24 Jan 2022]. Available from: https://clinicaltrials.gov/ct2/show/NCT03604705.
  • Shaw KJ, Ibrahim AS. Fosmanogepix: a review of the first-in-class broad spectrum agent for the treatment of invasive fungal infections. J Fungi. 2020;6(4):239.
  • Pfaller MA, Huband MD, Flamm RK, et al. Antimicrobial activity of manogepix, a first-in-class antifungal, and comparator agents tested against contemporary invasive fungal isolates from an international surveillance programme (2018–2019). J Glob Antimicrob Res. 2021;26:117–127.
  • Miyazaki M, Horii T, and Hata K, et al. In vitro activity of E1210, a novel antifungal, against clinically important yeasts and molds. Antimicrob Agents Chemother. 2011;55(10):4652–4658.
  • Watanabe NA, Miyazaki M, Horii T, et al. E1210, a new broad-spectrum antifungal, suppresses Candida albicans hyphal growth through inhibition of glycosylphosphatidylinositol biosynthesis. Antimicrob Agents Chemother. 2012;56(2):960–971.
  • Ibrexafungerp, DrugBank. [cited 24 Jan 2022]. Available from: https://go.drugbank.com/drugs/DB12471
  • SA W, Randolph R, Park S, et al. Preclinical pharmacokinetics and pharmacodynamic target of SCY-078, a first-in-class orally active antifungal glucan synthesis inhibitor, in murine models of disseminated candidiasis. Antimicrob Agents Chemother. 2017;61(4):AAC.02068–16.
  • Approved Drug FDA Products: brexafemme (ibrexafungerp) oral tablet: [cited 24 Jan 2022]. https://www.accessdata.fda.gov/drugsatfda_docs/label/2021/214900s000lbl.pdf
  • Ha YS, Covert SF, Momany M. FsFKS1, the 1,3-beta-glucan synthase from the caspofungin-resistant fungus Fusarium solani. Eukaryot Cell. 2006;5(7):1036–1042.
  • Perlin DS. Mechanisms of echinocandin antifungal drug resistance. Ann N Y Acad Sci. 2015;1354:1–11.
  • Billamboz M, Fatima Z, and Hameed S, et al., Promising drug candidates and new strategies for fighting against the emerging superbug Candida auris. Microorganisms. 2021;9(3): 634.
  • Rauseo AM, Coler-Reilly A, Larson L, et al. Hope on the horizon: novel fungal treatments in development. Open Forum Inf Dis. 2020;7(2):ofaa016.
  • Siramesine, DrugBank. [cited 24 Jan 2022]. Available from: https://go.drugbank.com/drugs/DB06555
  • Hanner M, Moebius FF, Flandorfer A, et al. Purification, molecular cloning, and expression of the mammalian sigma1-binding site. Proc Natl Acad Sci USA. 1996;93(15):8072–8077.
  • Vlainic J, Jovic O, and Kosalec I, et al. In vitro confirmation of siramesine as a novel antifungal agent with in silico lead proposals of structurally related antifungals. Molecules. 2021;26(12):3504.
  • Ebselen, DrugBank. [cited 24 Jan 2022]. Available from: https://go.drugbank.com/drugs/DB12610
  • May HC, Yu JJ, Guentzel MN, et al. Repurposing auranofin, ebselen, and PX-12 as antimicrobial agents targeting the thioredoxin system. Front Microbiol. 2018;9:336.
  • de Oliveira HC, Monteiro MC, and Rossi SA, et al. Identification of off-patent compounds that present antifungal activity against the emerging fungal pathogen Candida auris. Front Cell Infect Microbiol. 2019;9:83.
  • Marshall AC, Kidd SE, and Lamont-Friedrich SJ, et al. Structure, mechanism, and inhibition of Aspergillus fumigatus thioredoxin reductase. Antimicrob Agents Chemother. 2019;63(3):e02281–18.
  • Ngo HX, Shrestha SK, Garneau-Tsodikova S. Identification of ebsulfur analogues with broad-spectrum antifungal activity. ChemMedChem. 2016;11:1507–1516.
  • Wall G, Chaturvedi AK, and Wormley FL, et al. 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:e01084–18.
  • Plackett B. Why big pharma has abandoned antibiotics. Nature. 2020;586:S50–2.
  • Boyd NK, Teng C, Frei CR. Brief overview of approaches and challenges in new antibiotic development: a focus on drug repurposing. Front Cell Infect Microbiol. 2021;11:684515.
  • Schcolnik-Cabrera A, Juarez-Lopez D, Duenas-Gonzalez A. Perspectives on drug repurposing. Curr Med Chem. 2021;28(11):2085–2099.
  • Repurposing Drugs, National center for advancing translational sciences, NIH, Bethesda, MD, USA, 2022. [cited 10 Jan 2022]. Available from: https://ncats.nih.gov/preclinical/repurpose
  • Brown D. Antibiotic resistance breakers: can repurposed drugs fill the antibiotic discovery void? Nat Rev Drug Discov. 2015;14(12):821–832.
  • Farha MA, Brown ED. Drug repurposing for antimicrobial discovery. Nat Microbiol. 2019;4:565–577.
  • Miro-Canturri A, Ayerbe-Algaba R, Smani Y. Drug repurposing for the treatment of bacterial and fungal infections. Front Microbiol. 2019;10:41.
  • Pacios O, Blasco L, Bleriot I, et al., Strategies to combat multidrug-resistant and persistent infectious diseases. Antibiotics. 9(2): 65. 2020.
  • Kim JH, Cheng LW, Chan KL, et al., Antifungal drug repurposing. Antibiotics. 2020;9(11): 812.
  • Peyclit L, Yousfi H, Rolain JM, et al. Drug repurposing in medical mycology: identification of compounds as potential antifungals to overcome the emergence of multidrug-resistant fungi. Pharmaceuticals. 2021;14(5):488.
  • Zhang Q, Liu F, Zeng M, et al., Drug repurposing strategies in the development of potential antifungal agents. Appl Microbiol Biotechnol. 2021;105(13): 5259–5279.
  • Boyd NK, Teng C, Frei CR. Brief overview of approaches and challenges in new antibiotic development: a focus on drug repurposing. Front Cell Infect Microbiol. 2021;11:684515.
  • Siles SA, Srinivasan A, Pierce CG, et al. 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.
  • You Z, Ran X, Dai Y, et al. Clioquinol, an alternative antimicrobial agent against common pathogenic microbe. J Mycol Med. 2018;28:492–501.
  • Ortiz SC, Huang M, Hull CM. Spore germination as a target for antifungal therapeutics. Antimicrob Agents Chemother. 2019;63(12):e00994–19.
  • Wall G, Herrera N, Lopez-Ribot JL. Repositionable compounds with antifungal activity against multidrug resistant Candida auris identified in the medicines for malaria venture’s pathogen box. J Fungi. 2019;5(4):92.
  • Leonardelli F, Macedo D, and Dudiuk C, et al. In vitro activity of combinations of zinc chelators with amphotericin B and posaconazole against six mucorales species. Antimicrob Agents Chemother. 2019;63(5):e00266–19.
  • Coelho RA, Joffe LS, Alves GM, et al. A screening of the MMV Pathogen Box® reveals new potential antifungal drugs against the etiologic agents of chromoblastomycosis. PLoS One. 2020;15:e0229630.
  • Yousfi H, Ranque S, Cassagne C, et al. Identification of repositionable drugs with novel antimycotic activity by screening the Prestwick chemical library against emerging invasive moulds. J Glob Antimicrob Resist. 2020;21:314–317.
  • Cheng YS, Roma JS, and Shen M, et al. Identification of antifungal compounds against multidrug-resistant Candida auris utilizing a high-throughput drug-repurposing screen. Antimicrob Agents Chemother. 2021;65(4):e01305–20.
  • Almeida-Paes R, de Andrade IB, Ramos MLM, et al. Medicines for malaria venture COVID Box: a source for repurposing drugs with antifungal activity against human pathogenic fungi. Mem Inst Oswaldo Cruz. 2021;116:e210207.
  • Eldesouky HE, Mayhoub A, Hazbun TR, et al. Reversal of azole resistance in Candida albicans by sulfa antibacterial drugs. Antimicrob Agents Chemother. 2018;62(3):e00701–17.
  • Caldara M, Marmiroli N. Tricyclic antidepressants inhibit Candida albicans growth and biofilm formation. Int J Antimicrob Agents. 2018;52(4):500–505.
  • Iatta R, Puttilli MR, and Immediato D, et al. The role of drug efflux pumps in Malassezia pachydermatis and Malassezia furfur defence against azoles. Mycoses. 2017;60(3):178–182.
  • Aneke CI, Rhimi W, and Otranto D, et al. Synergistic effects of efflux pump modulators on the azole antifungal susceptibility of Microsporum canis. Mycopathologia. 2020;185(2):279–288.
  • Bulatova NR, Darwish RM. Effect of chemosensitizers on minimum inhibitory concentrations of fluconazole in Candida albicans. Med Princ Pract. 2008;17(2):117–121.
  • Homa M, Galgoczy L, and Toth E, et al. In vitro antifungal activity of antipsychotic drugs and their combinations with conventional antifungals against Scedosporium and Pseudallescheria isolates. Med Mycol. 2015;53(8):890–895.
  • Montoya MC, DiDone L, Heier RF, et al. Antifungal phenothiazines: optimization, characterization of mechanism, and modulation of neuroreceptor activity. ACS Infect Dis. 2018;4(4):499–507.
  • Rossi SA, de Oliveira HC, and Agreda-Mellon D, et al. Identification of off-patent drugs that show synergism with amphotericin B or that present antifungal action against Cryptococcus neoformans and Candida spp. Antimicrob Agents Chemother. 2020;64(4):e01921–19.
  • Galgoczy L, Papp T, and Kovacs L, et al. In vitro activity of phenothiazines and their combinations with amphotericin B against Zygomycetes causing rhinocerebral zygomycosis. Med Mycol. 2009;47(3):331–335.
  • Spitzer M, Griffiths E, Blakely KM, et al. Cross-species discovery of syncretic drug combinations that potentiate the antifungal fluconazole. Mol Syst Biol. 2011;7:499.
  • Siavoshi F, Tavakolian A, and Foroumadi A, et al. Comparison of the effect of non-antifungal and antifungal agents on Candida isolates from the gastrointestinal tract. Arch Iran Med. 2012;15(1):27–31.
  • Stylianou M, Kulesskiy E, Lopes JP, et al. Antifungal application of nonantifungal drugs. Antimicrob Agents Chemother. 2014;58:1055–1062.
  • Ji C, Liu N, Tu J, et al. Drug repurposing of haloperidol: discovery of new benzocyclane derivatives as potent antifungal agents against cryptococcosis and candidiasis. ACS Infect Dis. 2020;6:768–786.
  • Holbrook SYL, Garzan A, Dennis EK, et al. Repurposing antipsychotic drugs into antifungal agents: synergistic combinations of azoles and bromperidol derivatives in the treatment of various fungal infections. Eur J Med Chem. 2017;139:12–21.
  • Rajasekharan SK, Lee JH, Lee J. Aripiprazole repurposed as an inhibitor of biofilm formation and sterol biosynthesis in multidrug-resistant Candida albicans. Int J Antimicrob Agents. 2019;54(4):518–523.
  • Zhai B, Wu C, Wang L, et al. The antidepressant sertraline provides a promising therapeutic option for neurotropic cryptococcal infections. Antimicrob Agents Chemother. 2012;56:3758–3766.
  • Cong L, Liao Y, and Yang S, et al. In vitro antifungal activity of sertraline and synergistic effects in combination with antifungal drugs against planktonic forms and biofilms of clinical Trichosporon asahii isolates. PLoS One. 2016;11(12):e0167903.
  • Villanueva-Lozano H, Gonzalez GM, Espinosa-Mora JE, et al. Evaluation of the expanding spectrum of sertraline against uncommon fungal pathogens. J Infect Chemother. 2020;26(3):309–311.
  • Iyer KR, Revie NM, Fu C, et al. Treatment strategies for cryptococcal infection: challenges, advances and future outlook. Nat Rev Microbiol. 2021;19(7):454–466.
  • Rodrigues AG, Araujo R, and Pina-Vaz C. Interaction of local anaesthetics with other antifungal agents against pathogenic Aspergillus. Int J Antimicrob Agents. 2006;27(4):339–343.
  • Bagar T, and Bencina M. Antiarrhythmic drug amiodarone displays antifungal activity, induces irregular calcium response and intracellular acidification of Aspergillus niger - amiodarone targets calcium and pH homeostasis of A. niger. Fungal Genet Biol. 2012;49(10):779–791.
  • Liu S, Yue L, Gu W, et al. Synergistic effect of fluconazole and calcium channel blockers against resistant Candida albicans. PLoS One. 2016;11(3):e0150859.
  • Truong M, Monahan LG, Carter DA, et al. Repurposing drugs to fast-track therapeutic agents for the treatment of cryptococcosis. Peer J. 2018;6:e4761.
  • Alnajjar LM, Bulatova NR, and Darwish RM. Evaluation of four calcium channel blockers as fluconazole resistance inhibitors in Candida glabrata. J Glob Antimicrob Resist. 2018;14:185–189.
  • Westermeyer C, and Macreadie IG. Simvastatin reduces ergosterol levels, inhibits growth and causes loss of mtDNA in Candida glabrata. FEMS Yeast Res. 2007;7(3):436–441.
  • Nyilasi I, Kocsube S, and Krizsan K, et al. In vitro synergistic interactions of the effects of various statins and azoles against some clinically important fungi. FEMS Microbiol Lett. 2010;307(2):175–184.
  • Nyilasi I, Kocsube S, Krizsan K, et al. Susceptibility of clinically important dermatophytes against statins and different statin-antifungal combinations. Med Mycol. 2014;52(2):140–148.
  • Silva THS, Araujo CV, Santos KMDC, et al. Synergic effect of simvastatin in combination with amphotericin B against environmental strains of Cryptococcus neoformans from northeastern Brazil: a prospective experimental study. Sao Paulo Med J. 2020;138(1):40–46.
  • Chamilos G, Lewis RE, Kontoyiannis DP. Lovastatin has significant activity against zygomycetes and interacts synergistically with voriconazole. Antimicrob Agents Chemother. 2006;50(1):96–103.
  • Zhou Y, Yang H, Zhou X, et al. Lovastatin synergizes with itraconazole against planktonic cells and biofilms of Candida albicans through the regulation on ergosterol biosynthesis pathway. Appl Microbiol Biotechnol. 2018;102(12):5255–5264.
  • Naeimi Eshkaleti M, Kordbacheh P, and Hashemi SJ, et al. In vitro activity of amphotericin B in combination with statins against clinical and environmental Rhizopus oryzae strains. Iran J Public Health. 2019;48(5):943–948.
  • Eldesouky HE, Salama EA, and Li X, et al. Repurposing approach identifies pitavastatin as a potent azole chemosensitizing agent effective against azole-resistant Candida species. Sci Rep. 2020;10(1):7525.
  • Lima WG, Alves-Nascimento LA, Andrade JT, et al. Are the statins promising antifungal agents against invasive candidiasis? Biomed Pharmacother. 2019;111:270–281.
  • Ribeiro NQ, Costa MC, Magalhaes TFF, et al. Atorvastatin as a promising anticryptococcal agent. Int J Antimicrob Agents. 2017;49:695–702.
  • Ajdidi A, Sheehan G, and Abu Elteen K, et al. Assessment of the in vitro and in vivo activity of atorvastatin against Candida albicans. J Med Microbiol. 2019;68(10):1497–1506.
  • Esfahani AN, Golestannejad Z, and Khozeimeh F, et al. Antifungal effect of atorvastatin against Candida species in comparison to fluconazole and nystatin. Med Pharm Rep. 2019;92(4):368–373.
  • Mahmoud DE, Faraag AHI, and Abu El-Wafa WM. In vitro study on the potential fungicidal effects of atorvastatin in combination with some azole drugs against multidrug resistant Candida albicans. World J Microbiol Biotechnol. 2021;37(11):191.
  • Alem MA, Douglas LJ. Effects of aspirin and other nonsteroidal anti-inflammatory drugs on biofilms and planktonic cells of Candida albicans. Antimicrob Agents Chemother. 2004;48(1):41–47.
  • Al-Janabi AA. Determination of antidermatophytic effects of non-steroidal anti-inflammatory drugs on Trichophyton mentagrophytes and Epidermophyton floccosum. Mycoses. 2011;54(5):e443–8.
  • Rosato A, Catalano A, and Carocci A, et al. In vitro interactions between anidulafungin and nonsteroidal anti-inflammatory drugs on biofilms of Candida spp. Bioorg Med Chem. 2016;24(5):1002–1005.
  • Ogundeji AO, Pohl CH, Sebolai OM. Repurposing of aspirin and ibuprofen as candidate anti-cryptococcus drugs. Antimicrob Agents Chemother. 2016;60(8):4799–4808.
  • Alves LR, Oliveira C, and Goldenberg S. Eukaryotic translation elongation factor-1 alpha is associated with a specific subset of mRNAs in Trypanosoma cruzi. BMC Microbiol. 2015;15:104.
  • Nargesi S, and Rezaie S. Investigation an antifungal activity of diclofenac sodium against hyphae formation in Aspergillus fumigatus with attention to the expression of Ef-1 gene. Iran J Public Health. 2018;47(5):770–772.
  • Yang S, Liao Y, and Cong L, et al. In vitro interactions between non-steroidal anti-inflammatory drugs and antifungal agents against planktonic and biofilm forms of Trichosporon asahii. PLoS One. 2016;11:e0157047.
  • Krol J, Nawrot U, Bartoszewicz M. Anti-candidal activity of selected analgesic drugs used alone and in combination with fluconazole, itraconazole, voriconazole, posaconazole and isavuconazole. J Mycol Med. 2018;28(2):327–331.
  • Vallieres C, Singh N, Alexander C, et al. Repurposing nonantifungal approved drugs for synergistic targeting of fungal pathogens. ACS Infect Dis. 2020;6:2950–2958.
  • Borba-Santos LP, Nucci M, Ferreira-Pereira A, et al. Anti-sporothrix activity of ibuprofen combined with antifungal. Braz J Microbiol. 2021;52(1):101–106.
  • Pereira PA, Trindade BC, Secatto A, et al. Celecoxib improves host defense through prostaglandin inhibition during Histoplasma capsulatum infection. Mediators Inflamm. 2013;2013:950981.
  • Ma Y, Yang L, Liu X, et al. Development of celecoxib-derived antifungals for crop protection. Bioorg Chem. 2020;97:103670.
  • Mendoza SR, Zamith-Miranda D, Takacs T, et al., Complex and controversial roles of eicosanoids in fungal pathogenesis. J Fungi. 2021;7(4): 254.
  • Ko HT, Hsu LH, Yang SY, et al. Repurposing the thrombopoietin receptor agonist eltrombopag as an anticryptococcal agent. Med Mycol. 2020;58(4):493–504.
  • Singh S, Fatima Z, Ahmad K, et al. Repurposing of respiratory drug theophylline against Candida albicans: mechanistic insights unveil alterations in membrane properties and metabolic fitness. J Appl Microbiol. 2020;129(4):860–875.
  • Nile C, Falleni M, Cirasola D, et al. Repurposing pilocarpine hydrochloride for treatment of Candida albicans infections. mSphere. 2019;4(1):e00689–18.
  • Eldesouky HE, Lanman NA, and Hazbun TR, et al. Aprepitant, an antiemetic agent, interferes with metal ion homeostasis of Candida auris and displays potent synergistic interactions with azole drugs. Virulence. 2020;11(1):1466–1481.
  • Chavez-Dozal AA, Lown L, and Jahng M, et al. In vitro analysis of finasteride activity against Candida albicans urinary biofilm formation and filamentation. Antimicrob Agents Chemother. 2014;58(10):5855–5862.
  • Su S, Shi X, Xu W, et al. Antifungal activity and potential mechanism of panobinostat in combination with fluconazole against Candida albicans. Front Microbiol. 2020;11:1584.
  • Butts A, Koselny K, Chabrier-Rosello Y, et al. Estrogen receptor antagonists are anti-cryptococcal agents that directly bind EF hand proteins and synergize with fluconazole in vivo. mBio. 2014;5(1):e00765–13.
  • Zhang X, Fang Y, and Jaiseng W, et al. Characterization of tamoxifen as an antifungal agent using the yeast Schizosaccharomyces pombe model organism. Kobe J Med Sci. 2015;61(2):E54–63.
  • Hai TP, Van AD, Ngan NTT, et al. The combination of tamoxifen with amphotericin B, but not with fluconazole, has synergistic activity against the majority of clinical isolates of Cryptococcus neoformans. Mycoses. 2019;62:818–825.
  • Muthular M, Balsamo F, Passero P, et al. Effects of tamoxifen on periodontal disease and Candida albicans of patients with breast cancer and other pathologies. Future Microbiol. 2019;14:129–137.
  • Liu Q, Guo X, Jiang G, et al. NADPH-cytochrome P450 reductase ccr1 is a target of tamoxifen and participates in its antifungal activity via regulating cell wall integrity in fission yeast. Antimicrob Agents Chemother. 2020;64(9):e00079–20.
  • Ngan NTT, Thanh H, Le N, et al. An open label randomized controlled trial of tamoxifen combined with amphotericin B and fluconazole for cryptococcal meningitis. Elife. 2021;10:e68929.
  • Delattin N, De Brucker K, Vandamme K, et al. Repurposing as a means to increase the activity of amphotericin B and caspofungin against Candida albicans biofilms. J Antimicrob Chemother. 2014;69(4):1035–1044.
  • Sun W, Park Y-D, and Sugui JA, et al. Rapid identification of antifungal compounds against Exserohilum rostratum using high throughput drug repurposing screens. PLoS One. 2013;8(8):e70506.
  • Kubica TF, Denardi LB, and Azevedo MI, et al. Antifungal activities of tacrolimus in combination with antifungal agents against fluconazole-susceptible and fluconazole-resistant Trichosporon asahii isolates. Braz J Infect Dis. 2016;20(6):539–545.
  • Schwarz P, Schwarz PV, and Felske-Zech H, et al. In vitro interactions between isavuconazole and tacrolimus, cyclosporin A or sirolimus against Mucorales. J Antimicrob Chemother. 2019;74(7):1921–1927.
  • Li W, Zhang ZW, and Luo Y, et al. Molecular epidemiology, in vitro susceptibility and exoenzyme screening of Malassezia clinical isolates. J Med Microbiol. 2020;69(3):436–442.
  • Schwarz P, and Dannaoui E. In vitro interaction between isavuconazole and tacrolimus, cyclosporin A, or sirolimus against Aspergillus species. J Fungi. 2020;6(3):103.
  • Pandey N, Tripathi M, Gupta MK, et al. Overexpression of efflux pump transporter genes and mutations in ERG11 pave the way to fluconazole resistance in Candida tropicalis: a study from a North India region. J Glob Antimicrob Resist. 2020;22:374–378.
  • Heitman J, Movva NR, Hall MN. Targets for cell cycle arrest by the immunosuppressant rapamycin in yeast. Science. 1991;253(5022):905–909.
  • Bastidas RJ, Shertz CA, and Lee SC, et al. Rapamycin exerts antifungal activity in vitro and in vivo against Mucor circinelloides via FKBP12-dependent inhibition of Tor. Eukaryot Cell. 2012;11(3):270–281.
  • Cruz MC, Goldstein AL, Blankenship J, et al. Rapamycin and less immunosuppressive analogs are toxic to Candida albicans and Cryptococcus neoformans via FKBP12-dependent inhibition of TOR. J Antimicrob Agents Chemother. 2001;45(11):3162–3170.
  • Wall G, Chen E, and Hull MV, et al. Screening the CALIBR ReFRAME library in search for inhibitors of Candida auris biofilm formation. Front Cell Infect Microbiol. 2020;10:597931.
  • Thangamani S, Maland M, Mohammad H, et al. Repurposing approach identifies auranofin with broad spectrum antifungal activity that targets Mia40-Erv1 pathway. Front Cell Infect Microbiol. 2017;7:4.
  • Wiederhold NP, Patterson TF, and Srinivasan A, et al. Repurposing auranofin as an antifungal: in vitro activity against a variety of medically important fungi. Virulence. 2017;8(2):138–142.
  • She P, Liu Y, and Wang Y, et al. Antibiofilm efficacy of the gold compound auranofin on dual species biofilms of Staphylococcus aureus and Candida sp. J Appl Microbiol. 2020;128(1):88–101.
  • Shukla S, Sauna ZE, Prasad R, et al. Disulfiram is a potent modulator of multidrug transporter Cdr1p of Candida albicans. Biochem Biophys Res Commun. 2004;322:520–525.
  • Sauna ZE, Shukla S, Ambudkar SV. Disulfiram, an old drug with new potential therapeutic uses for human cancers and fungal infections. Mol Biosyst. 2005;1:127–134.
  • Yousfi H, Cassagne C, and Ranque S, et al. Repurposing of ribavirin as an adjunct therapy against invasive Candida strains in an in vitro study. Antimicrob Agents Chemother. 2019;63(10):e00263–19.
  • Zhang M, Yan H, Lu M, et al. Antifungal activity of ribavirin used alone or in combination with fluconazole against Candida albicans is mediated by reduced virulence. Int J Antimicrob Agents. 2020;55(1):105804.
  • Eldesouky HE, Salama EA, and Lanman NA, et al. Potent synergistic interactions between lopinavir and azole antifungal drugs against emerging multidrug-resistant Candida auris. Antimicrob Agents Chemother. 2020;65(1):e00684–20.
  • Vallieres C, Raulo R, Dickinson M, et al. Novel combinations of agents targeting translation that synergistically inhibit fungal pathogens. Front Microbiol. 2018;9:2355.
  • Shinde RB, Raut JS, Chauhan NM, et al. Chloroquine sensitizes biofilms of Candida albicans to antifungal azoles. Braz J Infect Dis. 2013;17:395–400.
  • Weber SM, Levitz SM, Harrison TS. Chloroquine and the fungal phagosome. Curr Opin Microbiol. 2000;3:349–353.
  • Montoya MC, Beattie S, Alden KM, et al. Derivatives of the antimalarial drug mefloquine are broad-spectrum antifungal molecules with activity against drug-resistant clinical isolates. Antimicrob Agents Chemother. 2020;64(3):e02331–19.
  • Kulkarny VV, Chavez-Dozal A, Rane HS, et al. Quinacrine inhibits Candida albicans growth and filamentation at neutral pH. Antimicrob Agents Chemother. 2014;58(12):7501–7509.
  • Harrison TS, Griffin GE, Levitz SM. Conditional lethality of the diprotic weak bases chloroquine and quinacrine against Cryptococcus neoformans. J Infect Dis. 2000;182(1):283–289.
  • Clark A, Hemmelgarn T, and Danziger-Isakov L, et al. Intravenous pentamidine for Pneumocystis carinii/jiroveci pneumonia prophylaxis in pediatric transplant patients. Pediatr Transplant. 2015;19(3):326–331.
  • Pozzebon Venturini T, Rossato L, and Chassot F, et al. In vitro synergistic combinations of pentamidine, polymyxin B, tigecycline and tobramycin with antifungal agents against Fusarium spp. J Med Microbiol. 2016;65(8):770–774.
  • Mei Y, Jiang T, Zou Y, et al. FDA approved drug library screening identifies robenidine as a repositionable antifungal. Front Microbiol. 2020;11:996.
  • Brilhante RS, Caetano EP, and Lima RA, et al. In vitro antifungal activity of miltefosine and levamisole: their impact on ergosterol biosynthesis and cell permeability of dimorphic fungi. J Appl Microbiol. 2015;119(4):962–969.
  • Spadari CC, Vila T, and Rozental S, et al. Miltefosine has a postantifungal effect and induces apoptosis in Cryptococcus yeasts. Antimicrob Agents Chemother. 2018;62(8):e00312–18.
  • Brilhante RSN, Silva MLQD, and Pereira VS, et al. Potassium iodide and miltefosine inhibit biofilms of Sporothrix schenckii species complex in yeast and filamentous forms. Med Mycol. 2019;57(6):764–772.
  • Wu Y, Totten M, and Memon W, et al. In vitro antifungal susceptibility of the emerging multidrug-resistant pathogen Candida auris to miltefosine alone and in combination with amphotericin B. Antimicrob Agents Chemother. 2020;64:e02063.
  • Garcia C, Burgain A, Chaillot J, et al. A phenotypic small-molecule screen identifies halogenated salicylanilides as inhibitors of fungal morphogenesis, biofilm formation and host cell invasion. Sci Rep. 2018;8(1):11559.
  • Dehdashti SJ, Abbott J, Nguyen DT, et al. A high-throughput screening assay for assessing the viability of Cryptococcus neoformans under nutrient starvation conditions. Anal Bioanal Chem. 2013;405(21):6823–6829.
  • Pic E, Burgain A, Sellam A. Repurposing the anthelminthic salicylanilide oxyclozanide against susceptible and clinical resistant Candida albicans strains. Med Mycol. 2019;57(3):387–390.
  • Joffe LS, Schneider R, Lopes W, et al. The anti-helminthic compound mebendazole has multiple antifungal effects against Cryptococcus neoformans. Front Microbiol. 2017;8:535.
  • Nixon GL, McEntee L, Johnson A, et al. Repurposing and reformulation of the antiparasitic agent flubendazole for treatment of cryptococcal meningoencephalitis, a neglected fungal disease. Antimicrob Agents Chemother. 2018;62(4):e01909–17.
  • Gao L, Sun Y, He C, et al. Synergy between pyrvinium pamoate and azoles against exophiala dermatitidis. Antimicrob Agents Chemother. 2018;62:e02361–17.
  • Del Poeta M, Schell WA, and Dykstra CC, et al. Structure-in vitro activity relationships of pentamidine analogues and dication-substituted bis-benzimidazoles as new antifungal agents. Antimicrob Agents Chemother. 1998;42(10):2495–2502.
  • Wiederhold NP. Review of T-2307, an investigational agent that causes collapse of fungal mitochondrial membrane potential. J Fungi. 2021;7(2):130.
  • Koselny K, Green J, Favazzo L, et al. 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(4):268–280.
  • Koselny K, Green J, and DiDone L, et al. The celecoxib derivative AR-12 has broad-spectrum antifungal activity in vitro and improves the activity of fluconazole in a murine model of cryptococcosis. Antimicrob Agents Chemother. 2016;60(12):7115.
  • Cruz MC, Goldstein AL, Blankenship JR, et al. Calcineurin is essential for survival during membrane stress in Candida albicans. EMBO J. 2002;21(4):546–559.
  • Juvvadi PR, Fox D, Bobay BG, et al. Harnessing calcineurin-FK506-FKBP12 crystal structures from invasive fungal pathogens to develop antifungal agents. Nat Commun. 2019;10(1):4275.
  • Gobeil SM, Bobay BG, Juvvadi PR, et al., Leveraging fungal and human calcineurin-inhibitor structures, biophysical data, and dynamics to design selective and nonimmunosuppressive FK506 analogs. mBio. 2021;12(6): e0300021.
  • Yun J, Lee DG. Role of potassium channels in chlorogenic acid-induced apoptotic volume decrease and cell cycle arrest in Candida albicans. Biochim Biophys Acta. 2017;1861:585–592.
  • Song J, Li R, and Jiang J. Copper homeostasis in Aspergillus fumigatus: opportunities for therapeutic development. Front Microbiol. 2019;10:774.
  • Simm C, May RC. Zinc and iron homeostasis: target-based drug screening as new route for antifungal drug development. Front Cell Infect Microbiol. 2019;9:181.
  • Albacar M, Sacka L, and Calafi C, et al. The toxic effects of Ppz1 overexpression involve Nha1-mediated deregulation of K+ and H+ homeostasis. J Fungi. 2021;7(12):1010.
  • Zhang C, Ren Y, and Gu H, et al., Calcineurin-mediated intracellular organelle calcium homeostasis is required for the survival of fungal pathogens upon extracellular calcium stimuli. Virulence. 2021;12(1):1091–1110.
  • Robinson JR, Isikhuemhen OS, Anike FN. Fungal-metal interactions: a review of toxicity and homeostasis. J Fungi. 2021;7(3):225.
  • Soares LW, Bailao AM, Soares CMA, et al. Zinc at the host-fungus interface: how to uptake the metal? J Fungi. 2020;6(4):305.
  • Wilson D. The role of zinc in the pathogenicity of human fungal pathogens. Adv Appl Microbiol. 2021;117:35–61.
  • Bellotti D, Miller A, and Rowinska-Zyrek M, et al. Zn2+ and Cu2+ binding to the extramembrane loop of Zrt2, a zinc transporter of Candida albicans. Biomolecules. 2022;12(1):121.
  • Li Y, Sun L, Lu C, et al. Promising antifungal targets against Candida albicans based on ion homeostasis. Front Cell Infect Microbiol. 2018;8:286.
  • Okoli I, Coleman JJ, Tampakakis E, et al. Identification of antifungal compounds active against Candida albicans using an improved high-throughput caenorhabditis elegans assay. PLoS One. 2009;4:e7025.
  • Martinez G, Regente M, Jacobi S, et al. Chlorogenic acid is a fungicide active against phytopathogenic fungi. Pestic Biochem Physiol. 2017;140:30–35.
  • Yun J, Lee DG. Role of potassium channels in chlorogenic acid-induced apoptotic volume decrease and cell cycle arrest in Candida albicans. Biochim Biophys Acta. 2017;1861:585–592.
  • Xu X, Zhao S, Yu Y, et al. A new antifungal beauvericin analogue from a marine-derived Fusarium sp. Nat Prod Commun. 2016;11(12):1825–1826.
  • Janeczko M, Kochanowicz E. Silymarin, a popular dietary supplement shows anti-candida activity. Antibiotics. 2019;8(4):206.
  • Singh S, Fatima Z, Hameed S. Insights into the mode of action of anticandidal herbal monoterpenoid geraniol reveal disruption of multiple MDR mechanisms and virulence attributes in Candida albicans. Arch Microbiol. 2016;198:459–472.
  • Gupta P, Gupta H, and Poluri KM. Geraniol eradicates Candida glabrata biofilm by targeting multiple cellular pathways. Appl Microbiol Biotechnol. 2021;105(13):5589–5605.
  • Cohrt KAO, Marin L, and Kjellerup L, et al. Novel zinc-attenuating compounds as potent broad-spectrum antifungal agents with in vitro and in vivo efficacy. Antimicrob Agents Chemother. 2018;62(5):e02024–17.
  • Moradian N, Ochs HD, Sedikies C, et al., The urgent need for integrated science to fight COVID-19 pandemic and beyond. J Transl Med. 2020;18(1): 205.
  • Badali H, Wiederhold NP. Antifungal resistance testing and implications for management. Curr Fungal Infect Rep. 2019;13:274–283.

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.