[en] The slowing-down de novo drug-discovery emphasized the importance of repurposing old drugs. This is particularly true when combating infections caused by therapy-refractory microorganisms, such as Scedosporium species and Lomentospora prolificans. Recent studies on Scedosporium responses to oxidative stress underscored the importance of targeting the underlying mechanisms. Auranofin, ebselen, PX-12, honokiol, and to a lesser extent, conoidin A are known to disturb redox-homeostasis systems in many organisms. Their antifungal activity was assessed against 27 isolates belonging to the major Scedosporium species: S. apiospermum, S. aurantiacum, S. boydii, S. dehoogii, S. minutisporum, and Lomentospora prolificans. Auranofin and honokiol were the most active against all Scedosporium species (mean MIC50 values of 2.875 and 6.143 μg/ml, respectively) and against L. prolificans isolates (mean MIC50 values of 4.0 and 3.563μg/ml respectively). Combinations of auranofin with voriconazole or honokiol revealed additive effects against 9/27 and 18/27 isolates, respectively. Synergistic interaction between auranofin and honokiol was only found against one isolate of L. prolificans. The effects of auranofin upon exposure to oxidative stress were also investigated. For all species except S. dehoogii, the maximal growth in the presence of auranofin significantly decreased when adding a sublethal dose of menadione. The analysis of the expression of genes encoding oxidoreductase enzymes upon exposure of S. apiospermum to honokiol unveiled the upregulation of many genes, especially those coding peroxiredoxins, thioredoxin reductases, and glutaredoxins. Altogether, these data suggest that auranofin and honokiol act via dampening the redox balance and support their repurposing as antifungals against Scedosporium species and L. prolificans.
Disciplines :
Veterinary medicine & animal health
Author, co-author :
Yaakoub, Hajar; Groupe d'Etude Des Interactions Hôte-Pathogène (GEIHP, EA 3142), SFR ICAT 4208, UNIV Angers, UNIV Brest, Institut De Biologie En Santé-IRIS, CHU Angers,Angers, France
Staerck, Cindy ; Université de Liège - ULiège > Fundamental and Applied Research for Animals and Health (FARAH) > FARAH: Santé publique vétérinaire ; Groupe d'Etude Des Interactions Hôte-Pathogène (GEIHP, EA 3142), SFR ICAT 4208, UNIV Angers, UNIV Brest, Institut De Biologie En Santé-IRIS, CHU Angers,Angers, France
Mina, Sara; Department of Medical Laboratory Sciences, Faculty of Health Sciences, Beirut Arab University, Beirut, Lebanon
Godon, Charlotte; Groupe d'Etude Des Interactions Hôte-Pathogène (GEIHP, EA 3142), SFR ICAT 4208, UNIV Angers, UNIV Brest, Institut De Biologie En Santé-IRIS, CHU Angers,Angers, France
Fleury, Maxime; Groupe d'Etude Des Interactions Hôte-Pathogène (GEIHP, EA 3142), SFR ICAT 4208, UNIV Angers, UNIV Brest, Institut De Biologie En Santé-IRIS, CHU Angers,Angers, France
Bouchara, Jean-Philippe; Groupe d'Etude Des Interactions Hôte-Pathogène (GEIHP, EA 3142), SFR ICAT 4208, UNIV Angers, UNIV Brest, Institut De Biologie En Santé-IRIS, CHU Angers,Angers, France ; Département de biologie des agents infectieux , Laboratoire De Parasitologie-Mycologie, Centre Hospitalier Universitaire, Angers, France
Calenda, Alphonse; Groupe d'Etude Des Interactions Hôte-Pathogène (GEIHP, EA 3142), SFR ICAT 4208, UNIV Angers, UNIV Brest, Institut De Biologie En Santé-IRIS, CHU Angers,Angers, France
Language :
English
Title :
Repurposing of auranofin and honokiol as antifungals against Scedosporium species and the related fungus Lomentospora prolificans.
MJJF, JPB and AC are members of the ECMM/ISHAM (European Confederation of Medical Mycology/International Society for Human and Animal Mycology) working group Fungal respiratory infections in Cystic Fibrosis (Fri-CF). For her PhD thesis, Cindy Staerck was recipient of a grant from our national patient organization against cystic fibrosis, Vaincre la Mucoviscidose, which is gratefully acknowledged. Some experiments were performed owing to the financial support from our local patient organization against cystic fibrosis Anjou Muco which is also acknowledged.
Ramirez-Garcia A, Pellon A, Rementeria A, et al. Scedosporium and Lomentospora: an updated overview of underrated opportunists. Med Mycol. 2018; 56 (suppl_1): 102–125.
Burgel PR, Bellis G, Olesen HV, et al. ERS/ECFS task force on provision of care for adults with cystic fibrosis in Europe. future trends in cystic fibrosis demography in 34 European countries. Eur Respir J. 2015; 46 (1): 133–141.
Bouchara JP, Le Govic Y, Kabbara S, et al. Advances in understanding and managing Scedosporium respiratory infections in patients with cystic fibrosis. Expert Rev Respir Med. 2020; 14 (3): 259–273.
Seidel D, Meißner A, Lackner M, et al. Prognostic factors in 264 adults with invasive Scedosporium spp. and Lomentospora prolificans infection reported in the literature and FungiScope®. Crit Rev Microbiol. 2019; 45 (1): 1–21.
Liu W, Feng RZ, Jiang H., Scedosporium spp lung infection in immunocompetent patients: a systematic review and MOOSE-compliant meta-analysis. Medicine (Baltimore). 2019; 98 (41): e17535.
Husain S, Muñoz P, Forrest G, et al. Infections due to Scedosporium apiospermum and Scedosporium prolificans in transplant recipients: clinical characteristics and impact of antifungal agent therapy on outcome. Clin Infect Dis. 2005; 40 (1): 89–99.
Parize P, Boussaud V, Poinsignon V, et al. Clinical outcome of cystic fibrosis patients colonized by Scedosporium species following lung transplantation: a single‐center 15‐year experience. Transplant Infect Dis. 2017; 19 (5): e12738.
Gilgado F, Serena C, Cano J, et al. Antifungal susceptibilities of the species of the Pseudallescheria boydii complex. Antimicrob Agents Chemother. 2006; 50 (12): 4211–4213.
Lackner M, De Hoog GS, Verweij PE, et al. Species-specific antifungal susceptibility patterns of Scedosporium and Pseudallescheria species. Antimicrob Agents Chemother. 2012; 56 (5): 2635–2642.
Staerck C, Vandeputte P, Gastebois A, et al. Enzymatic mechanisms involved in evasion of fungi to the oxidative stress: focus on Scedosporium apiospermum. Mycopathologia. 2018; 183 (1): 227–239.
Staerck C, Tabiasco J, Godon C, et al. Transcriptional profiling of Scedosporium apiospermum enzymatic antioxidant gene battery unravels the involvement of thioredoxin reductases against chemical and phagocytic cells oxidative stress. Med Mycol. 2019; 57 (3): 363–373.
Hirt RP, Müller S, Embley TM, et al. The diversity and evolution of thioredoxin reductase: new perspectives. Trends Parasitol. 2002; 18 (7): 302–308.
Martin-Vicente A, Guarro J, Capilla J., Does a triple combination have better activity than double combinations against multiresistant fungi? Experimental in vitro evaluation. Int J Antimicrob Agents. 2017; 49 (4): 422–426.
Schwarz C, Brandt C, Melichar V, et al. Combined antifungal therapy is superior to monotherapy in pulmonary scedosporiosis in cystic fibrosis. J Cyst Fibros. 2019; 18 (2): 227–232.
Jenks JD, Seidel D, Cornely OA, et al. Voriconazole plus terbinafine combination antifungal therapy for invasive Lomentospora prolificans infections: analysis of 41 patients from the fungiScope® registry 2008–2019. Clin Microbiol Infect. 2020; 26 (6): 784.e1-784.e5.
Pickering IJ, Cheng Q, Rengifo EM, et al. Direct observation of methylmercury and auranofin binding to selenocysteine in thioredoxin reductase. Inorg Chem. 2020; 59 (5): 2711–2718.
Harbut MB, Vilchèze C, Luo X, et al. Auranofin exerts broad-spectrum bactericidal activities by targeting thiol-redox homeostasis. Proc Natl Acad Sci U S A. 2015; 112 (14): 4453–4458.
Chen H, Liu Y, Liu Z, et al. Mutation in trxB leads to auranofin resistance in Staphylococcus aureus. J Glob Antimicrob Resist. 2020; 22: 135–136.
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.
Sharma N, Singh A, Sharma R, et al. Repurposing of auranofin against bacterial infections: an in silico and in vitro study. Curr Comput Aided Drug Des. 2020 Jul 17; 16. DOI: 10.2174/1386207323666200717155640.
Elkashif A, Seleem MN. Investigation of auranofin and gold-containing analogues antibacterial activity against multidrug-resistant Neisseria gonorrhoeae. Sci Rep. 2020; 10 (1): 5602.
Mohammad H, Abutaleb NS, Seleem MN. Auranofin rapidly eradicates methicillin-resistant Staphylococcus aureus (MRSA) in an infected pressure ulcer mouse model. Sci Rep. 2020; 10 (1): 7251.
She P, Liu Y, 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.
Feng L, Pomel S, Latre De Late P, et al. Repurposing auranofin and evaluation of a new gold (I) compound for the search of treatment of human and cattle parasitic diseases: from protozoa to helminth infections. Molecules. 2020; 25 (21): 5075.
Fuchs BB, RajaMuthiah R, Souza AC, et al. Inhibition of bacterial and fungal pathogens by the orphaned drug auranofin. Future Med Chem. 2016; 8 (2): 117–132.
Wiederhold NP, Patterson TF, 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.
Zembowicz A, Hatchett RJ, Radziszewski W, et al. Inhibition of endothelial nitric oxide synthase by ebselen. prevention by thiols suggests the inactivation by ebselen of a critical thiol essential for the catalytic activity of nitric oxide synthase. J Pharmacol Exp Ther. 1993; 267 (3): 1112–1118.
Schewe T. Molecular actions of ebselen—an antiinflammatory antioxidant. Gen Pharmacol. 1995; 26 (6): 1153–1169.
Thangamani S, Younis W, Seleem MN. Repurposing ebselen for treatment of multidrug-resistant staphylococcal infections. Sci Rep. 2015; 5 (1): 11596.
Thangamani S, Eldesouky HE, Mohammad H, et al. Ebselen exerts antifungal activity by regulating glutathione (GSH) and reactive oxygen species (ROS) production in fungal cells. Biochim Biophys Acta Gen Subj. 2017; 1861 (1): 3002–3010.
Ngo HX, Shrestha SK, Garneau‐Tsodikova S. Identification of Ebsulfur analogues with broad‐spectrum antifungal activity. ChemMedChem. 2016; 11 (14): 1507–1516.
Venturini TP, Chassot F, És L, et al. Antifungal activities of diphenyl diselenide and ebselen alone and in combination with antifungal agents against Fusarium spp. Med Mycol. 2016; 54 (5): 550–555.
Wall G, Chaturvedi AK, Wormley FLJ, 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 (10): e01084–18.
Carey KL, Westwood NJ, Mitchison TJ, et al. A small-molecule approach to studying invasive mechanisms of Toxoplasma gondii. Proc Natl Acad Sci U S A. 2004; 101 (19): 7433–7438.
Haraldsen JD, Liu G, Botting CH, et al. Identification of conoidin A as a covalent inhibitor of peroxiredoxin II. Org Biomol Chem. 2009; 7 (15): 3040–3048.
Nguyen JB, Pool CD, Wong CY, et al. Peroxiredoxin-1 from the human hookworm Ancylostoma ceylanicum forms a stable oxidized decamer and is covalently inhibited by conoidin A. Chem Biol. 2013; 20 (8): 991–1001.
Brizuela M, Huang HM, Smith C, et al. Treatment of erythrocytes with the 2-cys peroxiredoxin inhibitor, Conoidin A, prevents the growth of Plasmodium falciparum and enhances parasite sensitivity to chloroquine. PLoS One. 2014; 9 (4): e92411.
Kirkpatrick DL, Kuperus M, Dowdeswell M, et al. Mechanisms of inhibition of the thioredoxin growth factor system by antitumor 2-imidazolyl disulfides. Biochem Pharmacol. 1998; 55 (7): 987–994.
Ramanathan RK, Kirkpatrick DL, Belani CP, et al. A phase I pharmacokinetic and pharmacodynamic study of PX-12, a novel inhibitor of thioredoxin-1, in patients with advanced solid tumors. Clin Cancer Res. 2007; 13 (7): 2109–2114.
Ramanathan RK, Abbruzzese J, Dragovich T, et al. A randomized phase II study of PX-12, an inhibitor of thioredoxin in patients with advanced cancer of the pancreas following progression after a gemcitabine-containing combination. Cancer Chemother Pharmacol. 2011; 67 (3): 503–509.
Leal SM, Vareechon C, Cowden S, et al. Fungal antioxidant pathways promote survival against neutrophils during infection. J Clin Invest. 2012; 122 (7): 2482–2498.
Sun L, Liao K, Wang D. Honokiol induces superoxide production by targeting mitochondrial respiratory chain complex I in Candida albicans. PLoS One. 2017; 12 (8): e0184003.
Zhu X, Cai J, Zhou F, et al. Genome-wide screening of budding yeast with honokiol to associate mitochondrial function with lipid metabolism. Traffic. 2018; 19 (11): 867–878.
Sun LM, Liao K. Saccharomyces cerevisiae Hog1 MAP kinase pathway is activated in response to honokiol exposure. J Appl Microbiol. 2018; 124 (3): 754–763.
Day AM, Quinn J. Stress-activated protein kinases in human fungal pathogens. Front Cell Infect Microbiol. 2019; 9: 261.
Chen YH, Lu MH, Guo DS, et al. Antifungal effect of magnolol and honokiol from Magnolia officinalis on Alternaria alternata causing tobacco brown spot. Molecules. 2019; 24 (11): 2140.
Jin J, Guo N, Zhang J, et al. The synergy of honokiol and fluconazole against clinical isolates of azole-resistant Candida albicans. Lett Appl Microbiol. 2010; 51 (3): 351–357.
Zhan L, Peng X, Lin J, et al. Honokiol reduces fungal load, Toll-like receptor-2, and inflammatory cytokines in Aspergillus fumigatus keratitis. Invest Ophthalmol Vis Sci. 2020; 61 (4): 48.
Arendrup MC, Guinea J, Cuenca-Estrella M, et al. And the Subcommittee on Antifungal Susceptibility Testing (AFST) of the ESCMID European Committee for Antimicrobial Susceptibility Testing (EUCAST). EUCAST DEFINITIVE DOCUMENT E. DEF 9.3. Method for the determination of broth dilution minimum inhibitory concentrations of antifungal agents for conidia forming moulds, 2015.
Walsh TJ, Peter J, McGough DA, et al. Activities of amphotericin B and antifungal azoles alone and in combination against Pseudallescheria boydii. Antimicrob Agents Chemother. 1995; 39 (6): 1361–1364.
Rondeau S, Couderc L, Dominique S, et al. High frequency of voriconazole-related phototoxicity in cystic fibrosis patients. Eur Respir J. 2012; 39 (3): 782–784.
Levine MT, Chandrasekar PH. Adverse effects of voriconazole: over a decade of use. Clin Transplant. 2016; 30 (11): 1377–1386.
Sparkes T, Lemonovich TL, Infectious Diseases AST. Communitiy of Practice. Interactions between anti‐infective agents and immunosuppressants—Guidelines from the American Society of Transplantation Infectious Diseases Community of Practice. Clin Transplant. 2019; 33 (9): e13510.
Gottlieb NL. Pharmacology of auranofin: overview and update. Scand J Rheumatol Suppl. 1986; 63: 19–28.
Thangamani S, Mohammad H, Abushahba MF, et al. Antibacterial activity and mechanism of action of auranofin against multi-drug resistant bacterial pathogens. Sci Rep. 2016; 6 (1): 22571.
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.
Debnath A, Parsonage D, Andrade RM, et al. A high-throughput drug screen for Entamoeba histolytica identifies a new lead and target. Nat Med. 2012; 18 (6): 956–960.
Zhuang Q, Pan R, Liu X, et al. A validated ultra-HPLC–MS/MS method for determination of honokiol in human plasma and its application to a clinical pharmacokinetic study. Bioanalysis. 2019; 11 (11): 1085–1098.
Sarrica A, Kirika N, Romeo M, et al. Safety and toxicology of magnolol and honokiol. Planta Med. 2018; 84 (16): 1151–1164.
Syu WJ, Shen CC, Lu JJ, et al. Antimicrobial and Cytotoxic Activities of Neolignans fromMagnolia officinalis. Chem Biodivers. 2004; 1 (3): 530–537.
Jiang QQ, Fan LY, Yang GL, et al. Improved therapeutic effectiveness by combining liposomal honokiol with cisplatin in lung cancer model. BMC Cancer. 2008; 8 (1): 242.
Bang KH, Kim YK, Min BS, et al. Antifungal activity of magnolol and honokiol. Arch Pharm Res. 2000; 23 (1): 46–49.
Sun L, Liao K, Wang D. Effects of magnolol and honokiol on adhesion, yeast-hyphal transition, and formation of biofilm by Candida albicans. PLoS One. 2015; 10 (2): e0117695.
Wang Z, Shen Y. Antifungal compound honokiol triggers oxidative stress responsive signalling pathway and modulates central carbon metabolism. Mycology. 2016; 7 (3): 124–133.
Sun L, Liao K, Hang C, et al. Honokiol induces reactive oxygen species-mediated apoptosis in Candida albicans through mitochondrial dysfunction. PLoS One. 2017; 12 (2): e0172228.
Zhu X, Zou S, Li Y, et al. Transcriptomic analysis of Saccharomyces cerevisiae upon honokiol treatment. Res Microbiol. 2017; 168 (7): 626–635.
She P, Zhou L, Li S, et al. Synergistic microbicidal effect of auranofin and antibiotics against planktonic and biofilm-encased S. aureus and E. faecalis. Front Microbiol. 2019; 10: 2453.
Sun H, Zhang Q, Wang R, et al. Resensitizing carbapenem- and colistin-resistant bacteria to antibiotics using auranofin. Nat Commun. 2020; 11 (1): 5263.