Combination Therapy to Treat Fungal Biofilm-Based Infections
Abstract
:1. Introduction
2. Fungal Biofilm-Based Infections: Problem and Current Therapeutic Options
3. The Search for Synergistic Antibiofilm Combinations
3.1. Drug Interactions: Synergy
3.2. Screening for Novel Antibiofilm Combinations
4. The Mode of Action of Antibiofilm Combinations
4.1. Antibiofilm Combinations _targeting Virulence Factors
4.1.1. Combinations _targeting Biofilm-Specific Structures
4.1.2. _targeting the Activity or Secretion of Degradative Enzymes
4.1.3. Antibiofilm Combinations _targeting Adhesins
4.1.4. Modulation of Quorum Sensing by Antibiofilm Combinations
4.2. Antibiofilm Combinations _targeting Tolerance Mechanisms
4.2.1. Antibiofilm Combinations _targeting Drug Efflux Pumps
4.2.2. Cell Membranes or Sterol Biosynthesis Pathways as a _target for Antibiofilm Combinations
4.2.3. Combinations _targeting Stress Response Pathways
5. Discussion and Conclusions
Author Contributions
Funding
Acknowledgments
Conflicts of Interest
Abbreviations
AmB | Amphotericin B |
FICI | Fractional Inhibitory Concentration Index |
BEC-2 | Biofilm Eradication Concentration 2 |
BIC-2 | Biofilm Inhibition Concentration 2 |
RT-qPCR | Reverse transcriptional quantitative PCR |
Als | Agglutinin-like sequence |
Hwp | Hyphal wall protein |
eDNA | Extracellular DNA |
DNase | Deoxyribonuclease I |
Sap | Secreted aspartyl proteinase |
AC17 | 2-Adamantanamine |
Hsp90 | Heat shock protein 90 |
References
- Donlan, R.M. Biofilms: Microbial Life on Surfaces. Emerg. Infect. Dis. 2002, 8, 881–890. [Google Scholar] [CrossRef] [PubMed]
- Ramage, G.; Williams, C. The Clinical Importance of Fungal Biofilms. Adv. Appl. Microbiol. 2013, 84, 27–83. [Google Scholar] [CrossRef] [PubMed]
- Hall-Stoodley, L.; Costerton, J.W.; Stoodley, P. Bacterial biofilms: From the natural environment to infectious diseases. Nat. Rev. Microbiol. 2004, 2, 95–108. [Google Scholar] [CrossRef] [PubMed]
- Ramage, G.; Rajendran, R.; Sherry, L.; Williams, C. Fungal biofilm resistance. Int. J. Microbiol. 2012, 2012, 528521. [Google Scholar] [CrossRef]
- Liu, X.; Tang, B.; Gu, Q.; Yu, X. Elimination of the formation of biofilm in industrial pipes using enzyme cleaning technique. MethodsX 2014, 1, 130–136. [Google Scholar] [CrossRef]
- Lewis, K. Riddle of Biofilm Resistance. Antimicrob. Agents Chemother. 2001, 45, 999–1007. [Google Scholar] [CrossRef] [Green Version]
- Hawser, S.P.; Douglas, L.J. Resistance of Candida albicans Biofilms to Antifungal Agents In Vitro. Antimicrob. Agents Chemother. 1995, 39, 2128–2131. [Google Scholar] [CrossRef] [Green Version]
- Alhede, M.; Bjarnsholt, T.; Givskov, M.; Alhede, M. Pseudomonas aeruginosa biofilms: Mechanisms of immune evasion. Adv. Appl. Microbiol. 2014, 86, 1–40. [Google Scholar] [CrossRef]
- Johnson, C.J.; Cabezas-Olcoz, J.; Kernien, J.F.; Wang, S.X.; Beebe, D.J.; Huttenlocher, A.; Ansari, H.; Nett, J.E. The Extracellular Matrix of Candida albicans Biofilms Impairs Formation of Neutrophil Extracellular Traps. PLoS Pathog. 2016, 12, e1005884. [Google Scholar] [CrossRef] [Green Version]
- Muzny, C.A.; Schwebke, J.R. Biofilms: An Underappreciated Mechanism of Treatment Failure and Recurrence in Vaginal Infections. Clin. Infect. Dis. 2015, 61, 601–606. [Google Scholar] [CrossRef] [Green Version]
- Morgenstern, M.; Post, V.; Erichsen, C.; Hungerer, S.; Bühren, V.; Militz, M.; Richards, R.G.; Moriarty, T.F. Biofilm formation increases treatment failure in Staphylococcus epidermidis device-related osteomyelitis of the lower extremity in human patients. J. Orthop. Res. 2016, 34, 1905–1913. [Google Scholar] [CrossRef] [PubMed]
- Tapiainen, T.; Hanni, A.M.; Salo, J.; Ikäheimo, I.; Uhari, M. Escherichia coli biofilm formation and recurrences of urinary tract infections in children. Eur. J. Clin. Microbiol. Infect. Dis. 2014, 33, 111–115. [Google Scholar] [CrossRef] [PubMed]
- National Institutes of Health. Research on Microbial Biofilms; Report No. PA-03-047; National Institutes of Health: Bethesda, MD, USA, 2002. [Google Scholar]
- National Institutes of Health. Immunology of Biofilms; Report No. PA-06-537; National Institutes of Health: Bethesda, MD, USA, 2006. [Google Scholar]
- Shakibaie, M.R. Bacterial Biofilm and its Clinical Implications. Ann. Microbiol. Res. 2018, 2, 45–50. [Google Scholar] [CrossRef]
- Livengood, S.J.; Drew, R.H.; Perfect, J.R. Combination Therapy for Invasive Fungal Infections. Curr. Fungal Infect. Rep. 2020, 14, 40–49. [Google Scholar] [CrossRef]
- Bink, A.; Pellens, K.; Cammue, B.P.A.; Thevissen, K. Anti-Biofilm Strategies: How to Eradicate Candida Biofilms? Open Mycol. J. 2011, 5, 29–38. [Google Scholar] [CrossRef] [Green Version]
- Janbon, G.; Quintin, J.; Lanternier, F.; d’Enfert, C. Studying fungal pathogens of humans and fungal infections: Fungal diversity and diversity of approaches. Genes Immun. 2019, 20, 403–414. [Google Scholar] [CrossRef]
- Cauda, R. Candidaemia in Patients with an Inserted Medical Device. Drugs 2009, 69 (Suppl. 1), S33–S38. [Google Scholar] [CrossRef]
- Lebeaux, D.; Fernández-hidalgo, N.; Chauhan, A.; Lee, S.; Ghigo, J.; Almirante, B.; Beloin, C. Management of infections related to totally implantable venous-access ports: Challenges and perspectives. Lancet Infect. Dis. 2014, 14, 146–159. [Google Scholar] [CrossRef] [Green Version]
- Tumbarello, M.; Fiori, B.; Trecarichi, E.M.; Posteraro, P.; Losito, A.R.; De Luca, A.; Sanguinetti, M.; Fadda, G.; Cauda, R.; Posteraro, B. Risk Factors and Outcomes of Candidemia Caused by Biofilm-Forming Isolates in a Tertiary Care Hospital. PLoS ONE 2012, 7, e33705. [Google Scholar] [CrossRef] [Green Version]
- Rautemaa, R.; Ramage, G. Oral candidosis—Clinical challenges of a biofilm disease. Crit Rev. Microbiol 2011, 37, 328–336. [Google Scholar] [CrossRef]
- Alves, M.J.; Barreira, J.C.M.; Carvalho, I.; Trinta, L.; Perreira, L.; Ferreira, I.C.F.R.; Pintado, M. Propensity for biofilm formation by clinical isolates from urinary tract infections: Developing a multifactorial predictive model to improve antibiotherapy. J. Med. Microbiol. 2014, 63, 471–477. [Google Scholar] [CrossRef] [PubMed]
- Stylianou, M.; Kulesskiy, E.; Lopes, J.P.; Granlund, M.; Wennerberg, K.; Urban, C.F. Antifungal application of nonantifungal drugs. Antimicrob. Agents Chemother. 2014, 58, 1055–1062. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mccormick, A.; Loeffler, J.; Ebel, F. Aspergillus fumigatus: Contours of an opportunistic human pathogen. Cell. Microbiol. 2010, 12, 1535–1543. [Google Scholar] [CrossRef] [PubMed]
- Murakawa, G.J.; Harvell, J.D.; Lubitz, P.; Schnoll, S.; Lee, S.; Berger, T. Cutaneous aspergillosis and acquired immunodeficiency syndrome. Arch. Dermatol. 2000, 136, 365–369. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kwon-chung, K.J.; Fraser, J.A.; Doering, T.L.; Wang, Z.A.; Janbon, G.; Idnurm, A.; Bahn, Y. Cryptococcus neoformans and Cryptococcus gattii, the Etiologic Agents of Cryptococcosis. Cold Spring Harb Perspect. Med. 2014, 4, a019760. [Google Scholar] [CrossRef] [PubMed]
- Dora, J.M.; Kelbert, S.; Deutschendorf, C.; Cunha, V.S.; Aquino, V.R.; Pires dos Santos, R.; Zubaran Goldani, L. Cutaneous cryptococccosis due to Cryptococcus gattii in immunocompetent hosts: Case report and review. Mycopathologia 2006, 161, 235–238. [Google Scholar] [CrossRef] [PubMed]
- Shrestha, S.; Tuladhar, N.R.; Basnyat, S.; Acharya, G.P.; Shrestha, P.; Kumar, P. Prevalence of vaginitis among pregnant women attending Paropakar Maternity and Women’s Hospital, Thapathali, Kathmandu, Nepal. Nepal Med. Coll J. 2011, 13, 293–296. [Google Scholar] [PubMed]
- Abdul-Aziz, M.; Mahdy, M.A.K.; Abdul-Ghani, R.; Alhilali, N.A.; Al-Mujahed, L.K.A.; Alabsi, S.A.; Al-Shawish, F.A.M.; Alsarari, N.J.M.; Bamashmos, W.; Abdulwali, S.J.H.; et al. Bacterial vaginosis, vulvovaginal candidiasis and trichomonal vaginitis among reproductive-aged women seeking primary healthcare in Sana’a city, Yemen. BMC Infect. Dis. 2019, 19, 879. [Google Scholar] [CrossRef]
- Swidsinski, A.; Mendling, W.; Loening-Baucke, V.; Ladhoff, A.; Swidsinski, S.; Hale, L.P.; Lochs, H. Adherent biofilms in bacterial vaginosis. Obstet. Gynecol. 2005, 106, 1013–1023. [Google Scholar] [CrossRef] [Green Version]
- Hardy, L.; Cerca, N.; Jespers, V.; Vaneechoutte, M.; Crucitti, T. Bacterial biofilms in the vagina. Res. Microbiol. 2017, 168, 865–874. [Google Scholar] [CrossRef] [Green Version]
- Wu, X.; Zhang, S.; Li, H.; Shen, L.; Dong, C.; Sun, Y.; Chen, H.; Xu, B.; Zhuang, W.; Deighton, M.; et al. Biofilm Formation of Candida albicans Facilitates Fungal Infiltration and Persister Cell Formation in Vaginal Candidiasis. Front. Microbiol. 2020, 11, 1117. [Google Scholar] [CrossRef] [PubMed]
- Harriott, M.M.; Lilly, E.A.; Rodriguez, T.E.; Fidel, P.L.; Noverr, M.C. Candida albicans forms biofilms on the vaginal mucosa. Microbiology 2010, 156, 3635–3644. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sobel, J.D. Editorial Commentary: Vaginal Biofilm: Much Ado about Nothing, or a New Therapeutic Challenge? Clin. Infect. Dis. 2015, 61, 607–608. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Loussert, C.; Schmitt, C.; Prevost, M.C.; Balloy, V.; Fadel, E.; Philippe, B.; Kauffmann-Lacroix, C.; Latgé, J.P.; Beauvais, A. In vivo biofilm composition of Aspergillus fumigatus. Cell. Microbiol. 2010, 12, 405–410. [Google Scholar] [CrossRef]
- LaFleur, M.D.; Kumamoto, C.A.; Lewis, K. Candida albicans biofilms produce antifungal-tolerant persister cells. Antimicrob. Agents Chemother. 2006, 50, 3839–3846. [Google Scholar] [CrossRef] [Green Version]
- Yang, S.; Hay, I.D.; Cameron, D.R.; Speir, M.; Cui, B.; Su, F.; Peleg, A.Y.; Lithgow, T.; Deighton, M.A.; Qu, Y. Antibiotic regimen based on population analysis of residing persister cells eradicates Staphylococcus epidermidis biofilms. Sci. Rep. 2015, 5, 18578. [Google Scholar] [CrossRef] [Green Version]
- Qu, Y.; Daley, A.J.; Istivan, T.S.; Rouch, D.A.; Deighton, M.A. Densely adherent growth mode, rather than extracellular polymer substance matrix build-up ability, contributes to high resistance of Staphylococcus epidermidis biofilms to antibiotics-authors’ response. J. Antimicrob. Chemother. 2010, 65, 1405–1411. [Google Scholar] [CrossRef]
- Xu, H.; Sobue, T.; Thompson, A.; Xie, Z.; Poon, K.; Ricker, A.; Cervantes, J.; Diaz, P.I.; Dongari-Bagtzoglou, A. Streptococcal co-infection augments candida pathogenicity by amplifying the mucosal inflammatory response. Cell. Microbiol. 2014, 16, 214–231. [Google Scholar] [CrossRef]
- Zhou, Y.; Ying, F.; Jin, X.; Jin, J.; Li, S.; Hu, Y.; Yan, X.; Li, H.; Dong, Y.; Zhu, H. Candida albican-bacterial polymicrobial biofilms in recurrent vulvovaginal candidiasis contributes to the drug resistance of the disease: An assessment based on in vivo and in vitro assays. Int. J. Clin. Exp. Pathol. 2016, 9, 12057–12067. [Google Scholar]
- Kojic, E.M.; Darouiche, R.O. Candida Infections of Medical Devices. Clin. Microbiol. Rev. 2004, 17, 255–267. [Google Scholar] [CrossRef] [Green Version]
- Inácio, C.P.; de Araújo, P.S.R.; Brayner, F.A.; Alves, L.C.; Veras, D.L.; Neves, R.P. Invasive Candida tropicalis Infection Caused by Catheter Biofilm in a Patient with Tongue Cancer. Mycopathologia 2019, 184, 345–346. [Google Scholar] [CrossRef] [PubMed]
- Lai, M.-Y.; Hsu, J.-F.; Chu, S.-M.; Wu, I.-H.; Huang, H.-R.; Chiang, M.-C.; Fu, R.-H.; Tsai, M.-H. Risk Factors and Outcomes of Recurrent Candidemia in Children: Relapse or Re-Infection? J. Clin. Med. 2019, 8, 99. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chen, S.C.A.; Sorrell, T.C. Antifungal agents. Med. J. Aust. 2007, 187, 404–409. [Google Scholar] [CrossRef] [PubMed]
- Klepser, M.E.; Wolfe, E.J.; Jones, R.N.; Nightingale, C.H.; Pfaller, M.A. Antifungal pharmacodynamic characteristics of fluconazole and amphotericin B tested against Candida albicans. Antimicrob. Agents Chemother. 1997, 41, 1392–1395. [Google Scholar] [CrossRef] [Green Version]
- Mesa-Arango, A.C.; Trevijano-Contador, N.; Román, E.; Sánchez-Fresneda, R.; Casas, C.; Herrero, E.; Argüelles, J.C.; Pla, J.; Cuenca-Estrella, M.; Zaragoza, O. The production of reactive oxygen species is a universal action mechanism of amphotericin B against pathogenic yeasts and contributes to the fungicidal effect of this drug. Antimicrob. Agents Chemother. 2014, 58, 6627–6638. [Google Scholar] [CrossRef] [Green Version]
- Ghannoum, M.A.; Rice, L.B. Antifungal Agents: Mode of Action, Mechanisms of Resistance, and Correlation of These Mechanisms with Bacterial Resistance. Clin. Microbiol. Rev. 1999, 12, 501–517. [Google Scholar] [CrossRef] [Green Version]
- Odds, F.C.; Brown, A.J.P.; Gow, N.A.R. Antifungal agents: Mechanisms of action. Trends Microbiol. 2003, 11, 272–279. [Google Scholar] [CrossRef]
- Anderson, T.M.; Clay, M.C.; Cioffi, A.G.; Diaz, K.A.; Hisao, G.S.; Tuttle, M.D.; Nieuwkoop, A.J.; Comellas, G.; Maryum, N.; Wang, S.; et al. Amphotericin forms an extramembranous and fungicidal sterol sponge. Nat. Chem. Biol. 2014, 10, 400–406. [Google Scholar] [CrossRef]
- Grela, E.; Zdybicka-Barabas, A.; Pawlikowska-Pawlega, B.; Cytrynska, M.; Wlodarczyk, M.; Grudzinski, W.; Luchowski, R.; Gruszecki, W.I. Modes of the antibiotic activity of amphotericin B against Candida albicans. Sci. Rep. 2019, 9, 17029. [Google Scholar] [CrossRef] [Green Version]
- Sawaya, B.P.; Briggs, J.P.; Schnermann, J. Amphotericin B Nephrotoxicity: The Adverse Consequences of Altered Membrane Properties. J. Am. Soc. Nephrol. 1995, 6, 154–164. [Google Scholar]
- Sorkine, P.; Nagar, H.; Weinbroum, A.; Setton, A.; Israitel, E.; Scarlatt, A.; Silbiger, A.; Rudick, V.; Kluger, Y.; Halpern, P. Administration of amphotericin B in lipid emulsion decreases nephrotoxicity: Results of a prospective, randomized, controlled study in critically ill patients. Crit. Care Med. 1996, 24, 1311–1315. [Google Scholar] [CrossRef] [PubMed]
- Patel, G.P.; Crank, C.W.; Leikin, J.B. An Evaluation of Hepatotoxicity and Nephrotoxicity of Liposomal Amphotericin B (L-AMB). J. Med. Toxicol. 2011, 7, 12–15. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hitchcock, C.A.; Dickinson, K.; Brown, S.B.; Evans, E.G.V.; Adams, D.J. Interaction of azole antifungal antibiotics with cytochrome P-450- dependent 14a-sterol demethylase purified from Candida albicans. Biochem. J. 1990, 266, 475–480. [Google Scholar] [CrossRef] [PubMed]
- Sud, I.J.; Feingold, D.S. Mechanisms of Action of the Antimycotic Imidazoles. J. Investig. Dermatol. 1981, 76, 438–441. [Google Scholar] [CrossRef] [Green Version]
- Lepesheva, G.I.; Waterman, M.R. Sterol 14alpha-demethylase cytochrome P450 (CYP51), a P450 in all Biological Kingdoms. Biochim. Biophys. Acta 2007, 1770, 467–477. [Google Scholar] [CrossRef] [Green Version]
- Whaley, S.G.; Berkow, E.L.; Rybak, J.M.; Nishimoto, A.T.; Barker, K.S.; Rogers, P.D. Azole Antifungal Resistance in Candida albicans and Emerging Non- albicans Candida Species. Front. Microbiol. 2017, 7, 2173. [Google Scholar] [CrossRef] [Green Version]
- Lamfon, H.; Porter, S.R.; Mccullough, M.; Pratten, J. Susceptibility of Candida albicans biofilms grown in a constant depth film fermentor to chlorhexidine, fluconazole and miconazole: A longitudinal study. J. Antimicrob. Chemother. 2004, 53, 383–385. [Google Scholar] [CrossRef] [Green Version]
- Sherry, L.; Kean, R.; McKloud, E.; O’Donnell, L.E.; Metcalfe, R.; Jones, B.L.; Ramage, G. Biofilms Formed by Isolates from Patients Are Heterogeneous and Insensitive to Fluconazole. Antimicrob. Agents Chemother. 2017, 61, e01065-17. [Google Scholar] [CrossRef] [Green Version]
- Sawistowska-Schröder, E.T.; Kerridge, D.; Perry, H. Echinocandin inhibition of 1,3-beta-D-glucan synthase from Candida albicans. FEBS Lett. 1984, 173, 134–138. [Google Scholar] [CrossRef] [Green Version]
- Wagner, C.; Graninger, W.; Presterl, E.; Joukhadar, C. The echinocandins: Comparison of their pharmacokinetics, pharmacodynamics and clinical applications. Pharmacology 2006, 78, 161–177. [Google Scholar] [CrossRef]
- Klis, F.M.; De Groot, P.; Hellingwerf, K. Molecular organization of the cell wall of Candida albicans. Med. Mycol. 2001, 39 (Suppl. 1), S1–S8. [Google Scholar] [CrossRef] [Green Version]
- Bernard, M.; Latgé, J.P. Aspergillus fumigatus cell wall: Composition and biosynthesis. Med. Mycol. Suppl. 2001, 39 (Suppl. 1), S9–S17. [Google Scholar] [CrossRef] [Green Version]
- Zarnowski, R.; Westler, W.M.; Lacmbouh, G.A.; Marita, J.M.; Bothe, J.R.; Bernhardt, J.; Sahraoui, A.L.H.; Fontainei, J.; Sanchez, H.; Hatfeld, R.D.; et al. Novel entries in a fungal biofilm matrix encyclopedia. MBio 2014, 5, e01333-14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pound, M.W.; Townsend, M.L.; Drew, R.H. Echinocandin pharmacodynamics: Review and clinical implications. J. Antimicrob. Chemother. 2010, 65, 1108–1118. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Grover, N. Echinocandins: A ray of hope in antifungal drug therapy. Indian J. Pharmacol. 2010, 42, 9–11. [Google Scholar] [CrossRef] [Green Version]
- Maligie, M.A.; Selitrennikoff, C.P. Cryptococcus neoformans resistance to echinocandins: (1,3)β-glucan synthase activity is sensitive to echinocandins. Antimicrob. Agents Chemother. 2005, 49, 2851–2856. [Google Scholar] [CrossRef] [Green Version]
- Ryder, N.S.; Dupont, M.C. Inhibition of squalene epoxidase by allylamine antimycotic compounds. A comparative study of the fungal and mammalian enzymes. Biochem. J. 1985, 230, 765–770. [Google Scholar] [CrossRef] [Green Version]
- Nowosielski, M.; Hoffmann, M.; Wyrwicz, L.S.; Stepniak, P.; Plewczynski, D.M.; Lazniewski, M.; Ginalski, K.; Rychlewski, L. Detailed mechanism of squalene epoxidase inhibition by terbinafine. J. Chem. Inf. Model. 2011, 51, 455–462. [Google Scholar] [CrossRef]
- Hay, R. Therapy of skin, hair and nail fungal infections. J. Fungi 2018, 4, 99. [Google Scholar] [CrossRef] [Green Version]
- Dixon, D.M.; Polak, A. In vitro and in vivo Drug Studies with Three Agents of Central Nervous System Phaeohyphomycosis. Exp. Chemother. 1987, 33, 129–140. [Google Scholar] [CrossRef]
- Chen, S.; Shaohua, L.; Zhixiang, L.; Yan, W.; Yating, T.; Jiawen, L. Comparison of the Effects of Three Different Anti-fungus Drugs on Candida Albicans of Murine Vaginal Mucosa. J. Huazhong Univ. Sci. Technol. Med. Sci. 2007, 27, 209–212. [Google Scholar] [CrossRef] [PubMed]
- Ghannoum, M.A.; Long, L.; Kim, H.G.; Cirino, A.J.; Miller, A.R.; Mallefet, P. Efficacy of terbinafine compared to lanoconazole and luliconazole in the topical treatment of dermatophytosis in a guinea pig model. Med. Mycol. 2010, 48, 491–497. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fiori, B.; Posteraro, B.; Torelli, R.; Tumbarello, M.; Perlin, D.S.; Fadda, G.; Sanguinetti, M. In Vitro Activities of Anidulafungin and Other Antifungal Agents against Biofilms Formed by Clinical Isolates of Different Candida and Aspergillus Species. Antimicrob. Agents Chemother. 2011, 55, 3031–3035. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Uppuluri, P.; Srinivasan, A.; Ramasubramanian, A.; Lopez-ribot, J.L. Effects of Fluconazole, Amphotericin B, and Caspofungin on Candida albicans Biofilms under Conditions of Flow and on Biofilm Dispersion. Antimicrob. Agents Chemother. 2011, 55, 3591–3593. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kuhn, D.M.; George, T.; Chandra, J.; Mukherjee, P.K.; Ghannoum, M.A. Antifungal Susceptibility of Candida Biofilms: Unique Efficacy of Amphotericin B Lipid Formulations and Echinocandins. Antimicrob. Agents Chemother. 2002, 46, 1773–1780. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Perfect, J.R. The antifungal pipeline: A reality check. Nat. Rev. Drug Discov. 2017, 16, 603–616. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chandra, J.; Ghannoum, M.A. CD101, a Novel Echinocandin, Possesses Potent Antibiofilm Activity against Early and Mature Candida albicans Biofilms. Antimicrob. Agents Chemother. 2018, 62, e01750-17. [Google Scholar] [CrossRef] [Green Version]
- Hu, X.; Huang, Y.Y.; Wang, Y.; Wang, X.; Hamblin, M.R. Antimicrobial photodynamic therapy to control clinically relevant biofilm infections. Front. Microbiol. 2018, 9, 1299. [Google Scholar] [CrossRef] [Green Version]
- Güzel Tunccan, Ö.; Kalkanci, A.; Unal, E.A.; Abdulmajed, O.; Erdoğan, M.; Dizbay, M.; Çaglar, K. The in vitro effect of antimicrobial photodynamic therapy on Candida and Staphylococcus biofilms. Turkish J. Med. Sci. 2018, 48, 873–879. [Google Scholar] [CrossRef]
- Carmello, J.C.; Alves, F.; Basso, F.G.; de Souza Costa, C.A.; Tedesco, A.C.; Lucas Primo, F.; de Mima, E.G.O.; Pavarina, A.C. Antimicrobial photodynamic therapy reduces adhesion capacity and biofilm formation of Candida albicans from induced oral candidiasis in mice. Photodiagnosis Photodyn. Ther. 2019, 27, 402–407. [Google Scholar] [CrossRef]
- Jordão, C.C.; Viana de Sousa, T.; Inêz Klein, M.; Mendonça Dias, L.; Pavarina, A.C.; Carmello, J.C. Antimicrobial photodynamic therapy reduces gene expression of Candida albicans in biofilms. Photodiagnosis Photodyn. Ther. 2020, 31, 101825. [Google Scholar] [CrossRef]
- Walraven, C.J.; Lee, S.A. Antifungal lock therapy. Antimicrob. Agents Chemother. 2013, 57, 1–8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Soliman, G.M. Nanoparticles as safe and effective delivery systems of antifungal agents: Achievements and challenges. Int. J. Pharm. 2017, 523, 15–32. [Google Scholar] [CrossRef] [PubMed]
- Thangamani, S.; Maland, M.; Mohammad, H.; Pascuzzi, P.E.; Avramova, L.; Koehler, C.M.; Hazbun, T.R.; Seleem, M.N. Repurposing approach identifies auranofin with broad spectrum antifungal activity that _targets Mia40-erv1 pathway. Front. Cell. Infect. Microbiol. 2017, 7, 4. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Madende, M.; Albertyn, J.; Sebolai, O.; Pohl, C.H. Caenorhabditis elegans as a model animal for investigating fungal pathogenesis. Med. Microbiol. Immunol. 2020, 209, 1–13. [Google Scholar] [CrossRef]
- Breger, J.; Fuchs, B.B.; Aperis, G.; Moy, T.I.; Ausubel, F.M.; Mylonakis, E. Antifungal chemical compounds identified using a C. elegans pathogenicity assay. PLoS Pathog. 2007, 3, e18. [Google Scholar] [CrossRef]
- Okoli, I.; Coleman, J.J.; Tempakakis, E.; An, W.F.; Holson, E.; Wagner, F.; Conery, A.L.; 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. [Google Scholar] [CrossRef] [Green Version]
- Ahamefule, C.S.; Qin, Q.; Odiba, A.S.; Li, S.; Moneke, A.N.; Ogbonna, J.C.; Jin, C.; Wang, B.; Fang, W. Caenorhabditis elegans-Based Aspergillus fumigatus Infection Model for Evaluating Pathogenicity and Drug Efficacy. Front. Cell. Infect. Microbiol. 2020, 10, 320. [Google Scholar] [CrossRef]
- Odds, F.C. Synergy, antagonism, and what the chequerboard puts between them. J. Antimicrob Chemother 2003, 52, 1. [Google Scholar] [CrossRef]
- De Cremer, K.; Lanckacker, E.; Cools, T.L.; Bax, M.; De Brucker, K.; Cos, P.; Cammue, B.P.A.; Thevissen, K. Artemisinins, New Miconazole Potentiators Resulting in Increased Activity against Candida albicans Biofilms. Antimicrob. Agents Chemother. 2015, 59, 421–426. [Google Scholar] [CrossRef] [Green Version]
- Katragkou, A.; Mccarthy, M.; Alexander, E.L.; Antachopoulos, C.; Meletiadis, J.; Jabra-rizk, M.A.; Petraitis, V.; Roilides, E.; Walsh, T.J. In vitro interactions between farnesol and fluconazole, amphotericin b or micafungin against candida albicans biofilms. J. Antimicrob. Chemother. 2015, 70, 470–478. [Google Scholar] [CrossRef] [PubMed]
- Bugli, F.; Posteraro, B.; Papi, M.; Torelli, R.; Maiorana, A.; Sterbini, F.P.; Posteraro, P.; Sanguinetti, M.; De Spirito, M. In vitro interaction between alginate lyase and amphotericin B against aspergillus fumigatus biofilm determined by different methods. Antimicrob. Agents Chemother. 2013, 57, 1275–1282. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kovács, R.; Bozó, A.; Gesztelyi, R.; Domán, M.; Kardos, G.; Nagy, F.; Tóth, Z.; Majoros, L. Effect of caspofungin and micafungin in combination with farnesol against Candida parapsilosis biofilms. Int. J. Antimicrob. Agents 2016, 47, 304–310. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Greco, W.R.; Bravo, G.; Parsons, J.C. The Search for Synergy: A Critical Review from a Response Surface Perspective. Pharmacol. Rev. 1995, 47, 331–385. [Google Scholar] [PubMed]
- Prichard, M.N.; Shipman, C. A three-dimensional model to analyze drug-drug interactions. Antivir. Res. 1990, 14, 181–206. [Google Scholar] [CrossRef] [Green Version]
- Meletiadis, J.; Verweij, P.E.; Te Dorsthorst, D.T.A.; Meis, J.F.G.M.; Mouton, J.W. Assessing in vitro combinations of antifungal drugs against yeasts and filamentous fungi: Comparison of different drug interaction models. Med. Mycol. 2005, 43, 133–152. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sun, S.; Li, Y.; Guo, Q.; Shi, C.; Yu, J.; Ma, L. In vitro interactions between tacrolimus and azoles against Candida albicans determined by different methods. Antimicrob. Agents Chemother. 2008, 52, 409–417. [Google Scholar] [CrossRef] [Green Version]
- Loewe, S. Die quantitativen Probleme der Pharmakologie. Ergeb. Physiol. 1928, 27, 47–187. [Google Scholar] [CrossRef]
- Zhao, W.; Sachsenmeier, K.; Zhang, L.; Sult, E.; Hollingsworth, R.E.; Yang, H. A new bliss independence model to analyze drug combination data. J. Biomol. Screen. 2014, 19, 817–821. [Google Scholar] [CrossRef] [Green Version]
- Bliss, C.I. The Toxicity of Poisons Applied Jointly. Ann. Appl. Biol. 1939, 26, 585–615. [Google Scholar] [CrossRef]
- Van Dijck, P.; Sjollema, J.; Cammue, B.P.A.; Lagrou, K.; Berman, J.; D’Enfert, C.; Andes, D.R.; Arendrup, M.C.; Brakhage, A.A.; Calderone, R.; et al. Methodologies for in vitro and in vivo evaluation of efficacy of antifungal and antibiofilm agents and surface coatings against fungal biofilms. Microb. Cell 2018, 5, 300–326. [Google Scholar] [CrossRef] [PubMed]
- Nguyen, M.H.; Barchiesi, F.; McGough, D.A.; Yu, V.L.; Rinaldi, M.G. In vitro evaluation of combination of fluconazole and flucytosine against Cryptococcus neoformans var. neoformans. Antimicrob. Agents Chemother. 1995, 39, 1691–1695. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhou, Y.; Wang, G.; Li, Y.; Liu, Y.; Song, Y.; Zheng, W.; Zhang, N.; Hu, X.; Yan, S.; Jia, J. In vitro interactions between aspirin and amphotericin B against planktonic cells and biofilm cells of Candida albicans and C. parapsilosis. Antimicrob. Agents Chemother. 2012, 56, 3250–3260. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ernst, E.J.; Klepser, M.E.; Pfaller, M.A. In vitro interaction of fluconazole and amphotericin B administered sequentially against Candida albicans: Effect of concentration and exposure time. Diagn. Microbiol. Infect. Dis. 1998, 32, 205–210. [Google Scholar] [CrossRef]
- Li, Y.; Chang, W.; Zhang, M.; Li, X.; Jiao, Y.; Lou, H. Synergistic and drug-resistant reversing effects of diorcinol D combined with fluconazole against Candida albicans. FEMS Yeast Res. 2015, 15, fov001. [Google Scholar] [CrossRef] [Green Version]
- Fernández-Rivero, M.E.; del Pozo, J.L.; Ramírez, P.; Valentín, E.; Ruiz-Gaitán, A.; Pemán, J.; Cantón, E. Time-kill assays of amphotericin B plus anidulafungin against Candida tropicalis biofilms formed on two different biomaterials. Int. J. Artif. Organs 2018, 41, 23–27. [Google Scholar] [CrossRef] [Green Version]
- Bauer, A.W.; Kirby, W.M.; Sherris, J.C.; Turck, M. Antibiotic susceptibility testing by a standardized single disk method. Am. J. Clin. Pathol. 1966, 45, 493–496. [Google Scholar] [CrossRef]
- Ziaei-Darounkalaei, N.; Ameri, M.; Zahraei-Salehi, T.; Ziaei-Darounkalaei, O.; Mohajer-Tabrizi, T.; Bornaei, L. AZDAST the new horizon in antimicrobial synergism detection. MethodsX 2016, 3, 43–52. [Google Scholar] [CrossRef]
- Rosenberg, A.; Ene, I.V.; Bibi, M.; Zakin, S.; Segal, E.S.; Ziv, N.; Dahan, A.M.; Colombo, A.L.; Bennett, R.J.; Berman, J. Antifungal tolerance is a subpopulation effect distinct from resistance and is associated with persistent candidemia. Nat. Commun. 2018, 9, 2470. [Google Scholar] [CrossRef] [Green Version]
- Sader, H.S.; Pignatari, A.C.C. E test: A novel technique for antimicrobial susceptibility testing. São Paulo Med. J. 1994, 112, 635–638. [Google Scholar] [CrossRef]
- Pankey, G.; Ashcraft, D.; Kahn, H.; Ismail, A. Time-kill assay and etest evaluation for synergy with polymyxin B and fluconazole against Candida glabrata. Antimicrob. Agents Chemother. 2014, 58, 5795–5800. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tu, B.; Yin, G.; Li, H. Synergistic effects of vorinostat (SAHA) and azoles against Aspergillus species and their biofilms. BMC Microbiol. 2020, 20, 28. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kong, Q.; Cao, Z.; Lv, N.; Zhang, H.; Liu, Y.; Hu, L.; Li, J. Minocycline and Fluconazole Have a Synergistic Effect Against Cryptococcus neoformans Both in vitro and in vivo. Front. Microbiol. 2020, 11, 836. [Google Scholar] [CrossRef] [PubMed]
- Delattin, N.; De brucker, K.; Vandamme, K.; Meert, E.; Marchand, A.; Chaltin, P.; Cammue, B.P.A.; 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. [Google Scholar] [CrossRef] [Green Version]
- Lafleur, M.D.; Sun, L.; Lister, I.; Keating, J.; Nantel, A.; Long, L.; Ghannoum, M.; North, J.; Lee, R.E.; Coleman, K.; et al. Potentiation of Azole Antifungals by 2-Adamantanamine. Antimicrob. Agents Chemother. 2013, 57, 3585–3592. [Google Scholar] [CrossRef] [Green Version]
- Tits, J.; Cools, F.; De Cremer, K.; De Brucker, K.; Berman, J.; Verbruggen, K.; Gevaert, B.; Cos, P.; Cammue, B.P.A.; Thevissen, K. Combination of Miconazole and Domiphen Bromide Is Fungicidal against Biofilms of Resistant Candida spp. Antimicrob. Agents Chemother. 2020, 64, e01296-20. [Google Scholar] [CrossRef]
- Chen, X.; Ren, B.; Chen, M.; Wang, Q.; Zhang, L.; Yan, G. NLLSS: Predicting Synergistic Drug Combinations Based on Semi-supervised Learning. PLoS Comput. Biol. 2016, 12, e1004975. [Google Scholar] [CrossRef] [Green Version]
- De Cremer, K.; De Brucker, K.; Staes, I.; Peeters, A.; Van Den Driessche, F.; Coenye, T.; Cammue, B.P.A.; Thevissen, K. Stimulation of superoxide production increases fungicidal action of miconazole against Candida albicans biofilms. Sci. Rep. 2016, 6, 27463. [Google Scholar] [CrossRef] [Green Version]
- Sekhon, B.S. Repositioning drugs and biologics: Re_targeting old/existing drugs for potential new therapeutic applications. J. Pharm. Educ. Res. 2013, 4, 1–15. [Google Scholar]
- Azevedo, M.M.; Teixeira-Santos, R.; Pilva, A.P.; Cruz, L.; Ricardo, E.; Pina-Vaz, C.; Rodrigues, A.G. The effect of antibacterial and non-antibacterial compounds alone or associated with antifugals upon fungi. Front. Microbiol. 2015, 6, 669. [Google Scholar] [CrossRef]
- Hubsher, G.; Haider, M.; Okun, M.S. Amantadine: The journey from fighting flu to treating Parkinson disease. Neurology 2012, 78, 1096–1099. [Google Scholar] [CrossRef] [PubMed]
- Qiang, L.; Guo, J.; Han, Y.; Jiang, J.; Su, X.; Liu, H.; Qi, Q.; Han, L. A novel anti Candida albicans drug screening system based on high-throughput microfluidic chips. Sci. Rep. 2019, 9, 8087. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chen, X.; Ren, B.; Chen, M.; Liu, M.X.; Ren, W.; Wang, Q.X.; Zhang, L.X.; Yan, G.Y. ASDCD: Antifungal Synergistic Drug Combination Database. PLoS ONE 2014, 9, e86499. [Google Scholar] [CrossRef] [PubMed]
- Zhou, Y.; Yang, H.; Zhou, X.; Luo, H.; Tang, F.; Yang, J.; Alterovitz, G.; Cheng, L.; Ren, B. Lovastatin synergizes with itraconazole against planktonic cells and biofilms of Candida albicans through the regulation on ergosterol biosynthesis pathway. Appl. Microbiol. Biotechnol. 2018, 102, 5255–5264. [Google Scholar] [CrossRef]
- Yu, Q.; Ding, X.; Xu, N.; Cheng, X.; Qian, K.; Zhang, B.; Xing, L.; Li, M. In vitro activity of verapamil alone and in combination with fluconazole or tunicamycin against Candida albicans biofilms. Int. J. Antimicrob. Agents 2013, 41, 179–182. [Google Scholar] [CrossRef]
- Winter, M.B.; Salcedo, E.C.; Lohse, M.B.; Hartooni, N.; Gulati, M.; Sanchez, H.; Takagi, J.; Hube, B.; Andes, D.R.; Johnson, A.D.; et al. Global identification of biofilm-specific proteolysis in Candida albicans. MBio 2016, 7, e01514-16. [Google Scholar] [CrossRef] [Green Version]
- Nobile, C.J.; Fox, E.P.; Nett, J.E.; Sorrells, T.R.; Mitrovich, Q.M.; Hernday, A.D.; Tuch, B.B.; Andes, D.R.; Johnson, A.D. A recently evolved transcriptional network controls biofilm development in Candida albicans. Cell 2012, 148, 126–138. [Google Scholar] [CrossRef] [Green Version]
- Casolari, C.; Rossi, T.; Baggio, G.; Coppi, A.; Zandomeneghi, G.; Ruberto, A.I.; Farina, C.; Fabio, G.; Zanca, A.; Castelli, M. Interaction between saquinavir and antimycotic drugs on C. albicans and C. neoformans strains. Pharmacol. Res. 2004, 50, 605–610. [Google Scholar] [CrossRef]
- Lohse, M.B.; Gulati, M.; Craik, C.S.; Johnson, A.D.; Nobile, C.J. Combination of Antifungal Drugs and Protease Inhibitors Prevent Candida albicans Biofilm Formation and Disrupt Mature Biofilms. Front. Microbiol. 2020, 11, 1027. [Google Scholar] [CrossRef]
- Scorzoni, L.; de Paula e Silva, A.C.; Marcos, C.M.; Assato, P.A.; de Melo, W.C.; de Oliveira, H.C.; Costa-Orlandi, C.B.; Mendes-giannini, M.J.S.; Fusco-almeida, A.M. Antifungal Therapy: New Advances in the Understanding and Treatment of Mycosis. Front. Microbiol. 2017, 8, 36. [Google Scholar] [CrossRef] [Green Version]
- Pippi, B.; Lopes, W.; Reginatto, P.; Silva, F.É.K.; Joaquim, A.R.; Alves, R.J.; Silveira, G.P.; Vainstein, M.H.; Andrade, S.F.; Fuentefria, A.M. New insights into the mechanism of antifungal action of 8-hydroxyquinolines. Saudi Pharm. J. 2019, 27, 41–48. [Google Scholar] [CrossRef] [PubMed]
- Nature Medicine. Mechanism matters. Nat. Med. 2010, 16, 347. [Google Scholar] [CrossRef] [PubMed]
- Rezaei, M.; Mehrabian, S.; Amini, K. The Synergistic Effect of Ketoconazole and Probiotic Bifidobacterium Bifidum on Isolated Composition of Expression of Candida Albicans Als Gene Biofilm from Oral Samples. Payavard 2020, 13, 429–438. [Google Scholar]
- Robbins, N.; Uppuluri, P.; Nett, J.; Rajendran, R.; Ramage, G.; Lopez-Ribot, J.L.; Andes, D.; Cowen, L.E. Hsp90 governs dispersion and drug resistance of fungal biofilms. PLoS Pathog. 2011, 7, e1002257. [Google Scholar] [CrossRef]
- Wang, T.; Shao, J.; Da, W.; Li, Q.; Shi, G.; Wu, D.; Wang, C. Strong Synergism of Palmatine and Fluconazole/Itraconazole Against Planktonic and Biofilm Cells of Candida Species and Efflux-Associated Antifungal Mechanism. Front. Microbiol. 2018, 9, 2892. [Google Scholar] [CrossRef]
- De Andrade Neto, J.B.; da Silva, C.R.; Barroso, F.D.; do Amaral Valente Sá, L.; de Sousa Campos, R.; S Aires do Nascimento, F.B.; Sampaio, L.S.; de Silva, A.R.; da Silva, L.J.; de Sá Carneiro, I.; et al. Synergistic effects of ketamine and azole derivatives on Candida spp. resistance to fluconazole. Future Microbiol. 2020, 15, 177–188. [Google Scholar] [CrossRef]
- Li, Y.; Jiao, P.; Li, Y.; Gong, Y.; Chen, X.; Sun, S. The Synergistic Antifungal Effect and Potential Mechanism of D-Penicillamine Combined With Fluconazole Against Candida albicans. Front. Microbiol. 2019, 10, 2853. [Google Scholar] [CrossRef] [Green Version]
- Gu, W.; Guo, D.; Zhang, L.; Xu, D.; Sun, S. The synergistic effect of azoles and fluoxetine against resistant Candida albicans strains is attributed to attenuating fungal virulence. Antimicrob. Agents Chemother. 2016, 60, 6179–6188. [Google Scholar] [CrossRef] [Green Version]
- Zhang, M.; Yan, H.; Lu, M.; Wang, D.; Sun, S. 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, 105804. [Google Scholar] [CrossRef]
- Liu, X.; Li, T.; Wang, D.; Yang, Y.; Sun, W.; Liu, J.; Sun, S. Synergistic antifungal effect of fluconazole combined with licofelone against resistant Candida albicans. Front. Microbiol. 2017, 8, 2101. [Google Scholar] [CrossRef]
- Singh, B.N.; Upreti, D.K.; Singh, B.R.; Pandey, G.; Verma, S.; Roy, S.; Naqvi, A.H.; Rawat, A.K.S. Quercetin sensitizes fluconazole-resistant Candida albicans to induce apoptotic cell death by modulating quorum sensing. Antimicrob. Agents Chemother. 2015, 59, 2153–2168. [Google Scholar] [CrossRef] [Green Version]
- Eldesouky, H.E.; Salama, E.A.; Li, X.; Hazbun, T.R.; Mayhoub, A.S.; Seleem, M.N. Repurposing approach identifies pitavastatin as a potent azole chemosensitizing agent effective against azole-resistant Candida species. Sci. Rep. 2020, 10, 7525. [Google Scholar] [CrossRef] [PubMed]
- Sun, W.; Wang, D.; Yu, C.; Huang, X.; Li, X.; Sun, S. Strong synergism of dexamethasone in combination with fluconazole against resistant Candida albicans mediated by inhibiting drug efflux and reducing virulence. Int. J. Antimicrob. Agents 2017, 50, 399–405. [Google Scholar] [CrossRef] [PubMed]
- Lu, M.; Yu, C.; Cui, X.; Shi, J.; Yuan, L.; Sun, S. Gentamicin synergises with azoles against drug-resistant Candida albicans. Int. J. Antimicrob. Agents 2018, 51, 107–114. [Google Scholar] [CrossRef]
- Li, X.; Yu, C.; Huang, X.; Sun, S. Synergistic effects and mechanisms of budesonide in combination with fluconazole against resistant candida albicans. PLoS ONE 2016, 11, e0168936. [Google Scholar] [CrossRef] [PubMed]
- Lu, M.; Yan, H.; Yu, C.; Yuan, L.; Sun, S. Proton pump inhibitors act synergistically with fluconazole against resistant Candida albicans. Sci. Rep. 2020, 10, 498. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liu, Y.; Ren, H.; Wang, D.; Zhang, M.; Sun, S.; Zhao, Y. The synergistic antifungal effects of gypenosides combined with fluconazole against resistant Candida albicans via inhibiting the drug efflux and biofilm formation. Biomed. Pharmacother. 2020, 130, 110580. [Google Scholar] [CrossRef]
- Shi, W.; Chen, Z.; Chen, X.; Cao, L.; Liu, P.; Sun, S. The combination of minocycline and fluconazole causes synergistic growth inhibition against Candida albicans: An in vitro interaction of antifungal and antibacterial agents. FEMS Yeast Res. 2010, 10, 885–893. [Google Scholar] [CrossRef] [Green Version]
- Jia, W.; Zhang, H.; Li, C.; Li, G.; Liu, X.; Wei, J. The calcineruin inhibitor cyclosporine a synergistically enhances the susceptibility of Candida albicans biofilms to fluconazole by multiple mechanisms. BMC Microbiol. 2016, 16, 113. [Google Scholar] [CrossRef]
- Uppuluri, P.; Nett, J.; Heitman, J.; Andes, D. Synergistic effect of calcineurin inhibitors and fluconazole against Candida albicans biofilms. Antimicrob. Agents Chemother. 2008, 52, 1127–1132. [Google Scholar] [CrossRef] [Green Version]
- Shekhar-Guturja, T.; Gunaherath, G.M.K.B.; Wijeratne, E.M.K.; Lambert, J.-P.; Averette, A.F.; Lee, S.C.; Kim, T.; Bahn, Y.-S.; Tripodi, F.; Ammar, R.; et al. Dual Action Antifungal Small Molecule Modulates Multidrug Efflux and TOR signaling. Nat. Chem. Biol. 2016, 12, 867–875. [Google Scholar] [CrossRef] [PubMed]
- Fernandes, K.E.; Weeks, K.; Carter, D.A. Lactoferrin is broadly active against yeasts and highly synergistic with amphotericin B. Antimicrob. Agents Chemother. 2020, 64, e02284-19. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Martins, M.; Henriques, M.; Lopez-Ribot, J.L.; Oliveira, R. Addition of DNase Improves the In Vitro Activity of Antifungal Drugs against Candida albicans Biofilms. Mycoses 2012, 55, 80–85. [Google Scholar] [CrossRef] [PubMed]
- Rajendran, R.; Williams, C.; Lappin, D.F.; Millington, O.; Martins, M.; Ramage, G. Extracellular DNA release acts as an antifungal resistance mechanism in mature Aspergillus fumigatus biofilms. Eukaryot. Cell 2013, 12, 420–429. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Khan, S.N.; Khan, S.; Misba, L.; Sharief, M.; Hashmi, A.; Khan, A.U. Synergistic fungicidal activity with low doses of eugenol and amphotericin B against Candida albicans. Biochem. Biophys. Res. Commun. 2019, 518, 459–464. [Google Scholar] [CrossRef] [PubMed]
- Garzon, A.C.; Amado, D.; Robert, E.; Parra Giraldo, C.M.; Le Pape, P. Impact of calmodulin inhibition by fluphenazine on susceptibility, biofilm formation and pathogenicity of caspofungin-resistant Candida glabrata. J. Antimicrob. Chemother. 2020, 75, 1187–1193. [Google Scholar] [CrossRef]
- Brunke, S.; Mogavero, S.; Kasper, L.; Hube, B. Virulence factors in fungal pathogens of man. Curr. Opin. Microbiol. 2016, 32, 89–95. [Google Scholar] [CrossRef]
- Casadevall, A.; Pirofski, L.A. Host-Pathogen Interactions: Redefining the Basic Concepts of Virulence and Pathogenicity. Infect. Immun. 1999, 67, 3703–3713. [Google Scholar] [CrossRef] [Green Version]
- Casadevall, A.; Pirofski, L.A. Host-pathogen interactions: The attributes of virulence. J. Infect. Dis. 2001, 184, 337–344. [Google Scholar] [CrossRef]
- Casadevall, A.; Pirofski, L.A. The damage-response framework of microbial pathogenesis. Nat. Rev. Microbiol. 2003, 1, 17–24. [Google Scholar] [CrossRef]
- Casadevall, A.; Pirofski, L.A. Virulence factors and their mechanisms of action: The view from a damage-response framework. J. Water Health 2009, 7 (Suppl. 1), S2–S18. [Google Scholar] [CrossRef]
- Jabra-Rizk, M.A.; Kong, E.F.; Tsui, C.; Nguyen, M.H.; Clancy, C.J.; Fidel, P.L.; Noverr, M. Candida albicans pathogenesis: Fitting within the host-microbe damage response framework. Infect. Immun. 2016, 84, 2724–2739. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zaragoza, O. Basic principles of the virulence of Cryptococcus. Virulence 2019, 10, 490–501. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Abad, A.; Victoria Fernández-Molina, J.; Bikandi, J.; Ramírez, A.; Margareto, J.; Sendino, J.; Luis Hernando, F.; Pontón, J.; Garaizar, J.; Rementeria, A. What makes Aspergillus fumigatus a successful pathogen? Genes and molecules involved in invasive aspergillosis. Rev. Iberoam. Micol. 2010, 27, 155–182. [Google Scholar] [CrossRef] [PubMed]
- Hogan, L.H.; Klein, B.S.; Levitz, S.M. Virulence factors of medically important fungi. Clin. Microbiol. Rev. 1996, 9, 469–488. [Google Scholar] [CrossRef] [PubMed]
- Rella, A.; Farnoud, A.M.; Del Poeta, M. Plasma membrane lipids and their role in fungal virulence. Prog. Lipid Res. 2016, 61, 63–72. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chandra, J.; Kuhn, D.M.; Mukherjee, P.K.; Hoyer, L.L.; Ghannoum, M.A. Biofilm Formation by the Fungal Pathogen Candida albicans: Development, Architecture, and Drug Resistance. J. Bacteriol. 2001, 183, 5385–5394. [Google Scholar] [CrossRef] [Green Version]
- Nobile, C.J.; Mitchell, A.P. Regulation of cell-surface genes and biofilm formation by the C. albicans transcription factor Bcr1p. Curr. Biol. 2005, 15, 1150–1155. [Google Scholar] [CrossRef] [Green Version]
- Liu, Y.; Filler, S.G. Candida albicans Als3, a multifunctional adhesin and invasin. Eukaryot. Cell 2011, 10, 168–173. [Google Scholar] [CrossRef] [Green Version]
- Nobile, C.J.; Nett, J.E.; Andes, D.R.; Mitchell, A.P. Function of Candida albicans adhesin hwp1 in biofilm formation. Eukaryot. Cell 2006, 5, 1604–1610. [Google Scholar] [CrossRef] [Green Version]
- Ramage, G.; VandeWalle, K.; López-Ribot, J.L.; Wickes, B.L. The filamentation pathway controlled by the Efg1 regulator protein is required for normal biofilm formation and development in Candida albicans. FEMS Microbiol. Lett. 2002, 214, 95–100. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Schweizer, A.; Rupp, S.; Taylor, B.N.; Röllinghoff, M.; Schröppel, K. The TEA/ATTS transcription factor CaTec1p regulates hyphal development and virulence in Candida albicans. Mol. Microbiol. 2000, 38, 435–445. [Google Scholar] [CrossRef] [PubMed]
- Sasani, E.; Khodavaisy, S.; Agha Kuchak Afshari, S.; Darabian, S.; Aala, F.; Rezaie, S. Pseudohyphae formation in Candida glabrata due to CO2 exposure. Curr. Med. Mycol. 2016, 2, 49–52. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jiang, C.; Li, Z.; Zhang, L.; Tian, Y.; Dong, D.; Peng, Y. Significance of hyphae formation in virulence of Candida tropicalis and transcriptomic analysis of hyphal cells. Microbiol. Res. 2016, 192, 65–72. [Google Scholar] [CrossRef]
- Pukkila-Worley, R.; Peleg, A.Y.; Tampakakis, E.; Mylonakis, E. Candida albicans hyphal formation and virulence assessed using a caenorhabditis elegans infection model. Eukaryot. Cell 2009, 8, 1750–1758. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Weerasekera, M.M.; Wijesinghe, G.K.; Jayarathna, T.A.; Gunasekara, C.P.; Fernando, N.; Kottegoda, N.; Samaranayake, L.P. Culture media profoundly affect Candida Albicans and Candida tropicalis growth, adhesion and biofilm development. Mem. Inst. Oswaldo Cruz 2016, 111, 697–702. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Haque, F.; Alfatah, M.; Ganesan, K.; Bhattacharyya, M.S. Inhibitory Effect of Sophorolipid on Candida albicans Biofilm Formation and Hyphal Growth. Sci. Rep. 2016, 6, 23575. [Google Scholar] [CrossRef] [Green Version]
- Al-fattani, M.A.; Douglas, L.J. Biofilm matrix of Candida albicans and Candida tropicalis: Chemical composition and role in drug resistance. J. Med. Microbiol. 2006, 55, 999–1008. [Google Scholar] [CrossRef]
- Mitchell, K.F.; Taff, H.T.; Cuevas, M.A.; Reinicke, E.L.; Sanchez, H.; Andes, D.R. Role of matrix β-1,3 glucan in antifungal resistance of non-albicans Candida biofilms. Antimicrob. Agents Chemother. 2013, 57, 1918–1920. [Google Scholar] [CrossRef] [Green Version]
- Kernien, J.F.; Snarr, B.D.; Sheppard, D.C.; Nett, J.E. The interface between fungal biofilms and innate immunity. Front. Immunol. 2018, 8, 1968. [Google Scholar] [CrossRef] [Green Version]
- Nett, J.E.; Zarnowski, R.; Cabezas-Olcoz, J.; Brooks, E.G.; Bernhardt, J.; Marchillo, K.; Mosher, D.F.; Andes, D.R. Host contributions to construction of three device-associated Candida albicans biofilms. Infect. Immun. 2015, 83, 4630–4638. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Shopova, I.; Bruns, S.; Thywissen, A.; Kniemeyer, O.; Brakhage, A.A.; Hillmann, F. Extrinsic extracellular DNA leads to biofilm formation and colocalizes with matrix polysaccharides in the human pathogenic fungus Aspergillus fumigatus. Front. Microbiol. 2013, 4, 141. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Reichhardt, C.; Ferreir, J.A.G.; Joubert, L.M.; Clemons, K.V.; Stevens, D.A.; Cegelski, L. Analysis of the Aspergillus fumigatus biofilm extracellular matrix by solid-state nuclear magnetic resonance spectroscopy. Eukaryot. Cell 2015, 14, 1064–1072. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nett, J.E.; Sanchez, H.; Cain, M.T.; Andes, D.R. Genetic basis of Candida Biofilm resistance due to drug-sequestering matrix glucan. J. Infect. Dis. 2010, 202, 171–175. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nett, J.E.; Crawford, K.; Marchillo, K.; Andes, D.R. Role of Fks1p and matrix glucan in Candida albicans biofilm resistance to an echinocandin, pyrimidine, and polyene. Antimicrob. Agents Chemother. 2010, 54, 3505–3508. [Google Scholar] [CrossRef] [Green Version]
- Nett, J.E.; Guite, K.M.; Ringeisen, A.; Holoyda, K.A.; Andes, D.R. Reduced biocide susceptibility in Candida albicans biofilms. Antimicrob. Agents Chemother. 2008, 52, 3411–3413. [Google Scholar] [CrossRef] [Green Version]
- Sheppard, D.C. Molecular Mechanisms of Aspergillus fumigatus Adherence to Host Constituents. Curr. Opin. Microbiol. 2011, 14, 375–379. [Google Scholar] [CrossRef] [Green Version]
- Gravelat, F.N.; Beauvais, A.; Liu, H.; Lee, M.J.; Snarr, B.D.; Chen, D.; Xu, W.; Kravtsov, I.; Hoareau, C.M.Q.; Vanier, G.; et al. Aspergillus Galactosaminogalactan Mediates Adherence to Host Constituents and Conceals Hyphal β-Glucan from the Immune System. PLoS Pathog. 2013, 9, e1003575. [Google Scholar] [CrossRef] [Green Version]
- Lee, M.J.; Geller, A.M.; Bamford, N.C.; Liu, H.; Gravelat, F.N.; Snarr, B.D.; Le Mauff, F.; Chabot, J.; Ralph, B.; Ostapska, H.; et al. Deacetylation of fungal exopolysaccharide mediates adhesion and biofilm formation. MBio 2016, 7, e00252-16. [Google Scholar] [CrossRef] [Green Version]
- Lee, M.J.; Liu, H.; Barker, B.M.; Snarr, B.D.; Gravelat, F.N.; Al Abdallah, Q.; Gavino, C.; Baistrocchi, S.R.; Ostapska, H.; Xiao, T.; et al. The Fungal Exopolysaccharide Galactosaminogalactan Mediates Virulence by Enhancing Resistance to Neutrophil Extracellular Traps. PLoS Pathog. 2015, 11, e1005187. [Google Scholar] [CrossRef]
- Martins, M.; Uppuluri, P.; Thomas, D.P.; Cleary, I.A.; Henriques, M.; Lopez-Ribot, J.L.; Oliveira, R. Presence of extracellular DNA in the Candida albicans biofilm matrix and its contribution to biofilms. Mycopathologia 2010, 169, 323–331. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Papi, M.; Maiorana, A.; Bugli, F.; Torelli, R.; Posteraro, B.; Maulucci, G.; De Spirito, M.; Sanguinetti, M. Detection of biofilm-grown Aspergillus fumigatus by means of atomic force spectroscopy: Ultrastructural effects of alginate lyase. Microsc. Microanal. 2012, 18, 1088–1094. [Google Scholar] [CrossRef] [PubMed]
- Karkowska-Kuleta, J.; Rapala-Kozik, M.; Kozik, A. Fungi pathogenic to humans: Molecular bases of virulence of Candida albicans, Cryptococcus neoformans and Aspergillus fumigatus. Acta Biochim. Pol. 2009, 56, 211–224. [Google Scholar] [CrossRef] [PubMed]
- Schaller, M.; Borelli, C.; Korting, H.C.; Hube, B. Hydrolytic enzymes as virulence factors of Candida albicans. Mycoses 2005, 48, 365–377. [Google Scholar] [CrossRef] [PubMed]
- Nailis, H.; Kucharíkov, S.; Řičicovńn, M.; Van Dijck, P.; Deforce, D.; Nelis, H.; Coenye, T. Real-time PCR expression profiling of genes encoding potential virulence factors in Candida albicans biofilms: Identification of model-dependent and -independent gene expression. BMC Microbiol. 2010, 10, 114. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Naglik, J.R.; Moyes, D.; Makwana, J.; Kanzaria, P.; Tsichlaki, E.; Weindl, G.; Tappuni, A.R.; Rodgers, C.A.; Woodman, A.J.; Challacombe, S.J.; et al. Quantitative expression of Candida albicans secreted aspartyl proteinase gene family in human oral and vaginal candidiasis. Microbiology. 2008, 154 Pt 11, 3266–3280. [Google Scholar] [CrossRef] [Green Version]
- Sacristán, B.; Blanco, M.T.; Galán-Ladero, M.A.; Blanco, J.; Pérez-Giraldo, C.; Gómez-Garca, A.C. Aspartyl proteinase, phospholipase, hemolytic activities and biofilm production of Candida albicans isolated from bronchial aspirates of ICU patients. Med. Mycol. 2011, 49, 94–97. [Google Scholar] [CrossRef] [Green Version]
- Rajendran, R.; Robertson, D.P.; Hodge, P.J.; Lappin, D.F.; Ramage, G. Hydrolytic Enzyme Production is Associated with Candida Albicans Biofilm Formation from Patients with Type 1 Diabetes. Mycopathologia 2010, 170, 229–235. [Google Scholar] [CrossRef]
- Schild, L.; Heyken, A.; de Groot, P.W.J.; Hiller, E.; Mock, M.; de Koster, C.; Horn, U.; Rupp, S.; Hube, B. Proteolytic cleavage of covalently linked cell wall proteins by Candida albicans Sap9 and Sap10. Eukaryot. Cell 2011, 10, 98–109. [Google Scholar] [CrossRef] [Green Version]
- Chaffin, W.L. Candida albicans Cell Wall Proteins. Microbiol. Mol. Biol. Rev. 2008, 72, 495–544. [Google Scholar] [CrossRef] [Green Version]
- Kadry, A.A.; El-Ganiny, A.M.; El-Baz, A.M. Relationship between Sap prevalence and biofilm formation among resistant clinical isolates of Candida albicans. Afr. Health Sci. 2018, 18, 1166–1174. [Google Scholar] [CrossRef] [PubMed]
- Ibrahim, A.S.; Mirbod, F.; Filler, S.G.; Banno, Y.; Cole, G.T.; Kitajima, Y.; Edwards, J.E.; Nozawa, Y.; Ghannoum, M.A. Evidence implicating phospholipase as a virulence factor of Candida albicans. Infect. Immun. 1995, 63, 1993–1998. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Price, M.F.; Wilkinson, I.D.; Gentry, L.O. Plate method for detection of phospholipase activity in Candida albicans. Sabouraudia 1982, 20, 7–14. [Google Scholar] [CrossRef] [PubMed]
- Naglik, J.R.; Challacombe, S.J.; Hube, B. Candida albicans Secreted Aspartyl Proteinases in Virulence and Pathogenesis. Microbiol. Mol. Biol. Rev. 2003, 67, 400–428. [Google Scholar] [CrossRef] [Green Version]
- De Groot, P.W.J.; Bader, O.; de Boer, A.D.; Weig, M.; Chauhan, N. Adhesins in human fungal pathogens: Glue with plenty of stick. Eukaryot. Cell 2013, 12, 470–481. [Google Scholar] [CrossRef] [Green Version]
- Hoyer, L.L.; Green, C.B.; Oh, S.H.; Zhao, X. Discovering the secrets of the Candida albicans agglutinin-like sequence (ALS) gene family—A sticky pursuit. Med. Mycol. 2008, 46, 1–15. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Albuquerque, P.; Casadevall, A. Quorum sensing in fungi—A review. Med. Mycol. 2012, 50, 337–345. [Google Scholar] [CrossRef] [Green Version]
- Hornby, J.M.; Jensen, E.C.; Lisec, A.D.; Tasto, J.J.; Shoemaker, R.; Dussault, P.; Nickerson, K.W. Quorum Sensing in the Dimorphic Fungus Candida albicans Is Mediated by Farnesol. Appl. Environ. Microbiol. 2001, 67, 2982–2992. [Google Scholar] [CrossRef] [Green Version]
- Ramage, G.; Saville, S.P.; Wickes, B.L.; López-ribot, J.L. Inhibition of Candida albicans Biofilm Formation by Farnesol, a Quorum-Sensing Molecule. Appl. Environ. Microbiol. 2002, 68, 5459–5463. [Google Scholar] [CrossRef] [Green Version]
- Langford, M.L.; Hargarten, J.C.; Patefield, K.D.; Marta, E.; Blankenship, J.R.; Fanning, S.; Nickerson, K.W.; Atkin, A.L. Candida albicans Czf1 and Efg1 coordinate the response to farnesol during quorum sensing, white-opaque thermal dimorphism, and cell death. Eukaryot. Cell 2013, 12, 1281–1292. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kalia, V.C. Quorum sensing inhibitors: An overview. Biotechnol. Adv. 2013, 31, 224–245. [Google Scholar] [CrossRef]
- Singh, B.N.; Singh, H.B.; Singh, A.; Singh, B.R.; Mishra, A.; Nautiyal, C.S. Lagerstroemia speciosa fruit extract modulates quorum sensing-controlled virulence factor Production and biofilm formation in Pseudomonas aeruginosa. Microbiology 2012, 158, 529–538. [Google Scholar] [CrossRef] [PubMed]
- Singh, B.N.; Singh, B.R.; Singh, R.L.; Prakash, D.; Dhakarey, R.; Upadhyay, G.; Singh, H.B. Oxidative DNA damage protective activity, antioxidant and anti-quorum sensing potentials of Moringa oleifera. Food Chem. Toxicol. 2009, 47, 1109–1116. [Google Scholar] [CrossRef] [PubMed]
- Adonizio, A.; Kong, K.F.; Mathee, K. Inhibition of quorum sensing-controlled virulence factor production in Pseudomonas aeruginosa by south Florida plant extracts. Antimicrob. Agents Chemother. 2008, 52, 198–203. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Singh, B.N.; Singh, B.R.; Singh, R.L.; Prakash, D.; Sarma, B.K.; Singh, H.B. Antioxidant and anti-quorum sensing activities of green pod of Acacia nilotica L. Food Chem. Toxicol. 2009, 47, 778–786. [Google Scholar] [CrossRef]
- Delarze, E.; Sanglard, D. Defining the frontiers between antifungal resistance, tolerance and the concept of persistence. Drug Resist. Updates 2015, 23, 12–19. [Google Scholar] [CrossRef] [Green Version]
- Berman, J.; Krysan, D.J. Drug resistance and tolerance in fungi. Nat. Rev. Microbiol. 2020, 18, 319–331. [Google Scholar] [CrossRef]
- Ramage, G.; Bachmann, S.; Patterson, T.F.; Wickes, B.L.; Lopez-Ribot, J.L. Investigation of multidrug efflux pumps in relation to fluconazole resistance in Candida albicans biofilms. J. Antimicrob. Chemother. 2002, 49, 973–980. [Google Scholar] [CrossRef] [Green Version]
- Mukherjee, P.K.; Chandra, J.; Kuhn, D.M.; Ghannoum, M.A. Mechanism of fluconazole resistance in Candida albicans biofilms: Phase-specific role of efflux pumps and membrane sterols. Infect. Immun. 2003, 71, 4333–4340. [Google Scholar] [CrossRef] [Green Version]
- Cannon, R.D.; Lamping, E.; Holmes, A.R.; Niimi, K.; Baret, P.V.; Keniya, M.V.; Tanabe, K.; Niimi, M.; Goffeau, A.; Monk, B.C. Efflux-mediated antifungal drug resistance. Clin. Microbiol. Rev. 2009, 22, 291–321. [Google Scholar] [CrossRef] [Green Version]
- Maesaki, S.; Marichal, P.; Vanden Bossche, H.; Sanglard, D.; Kohno, S. Rhodamine 6G efflux for the detection of CDR1-overexpressing azole-resistant Candida albicans strains. J. Antimicrob. Chemother. 1999, 44, 27–31. [Google Scholar] [CrossRef] [PubMed]
- Clark, F.S.; Parkinson, T.; Hitchcock, C.A.; Gow, N.A.R. Correlation between rhodamine 123 accumulation and azole sensitivity in Candida species: Possible role for drug efflux in drug resistance. Antimicrob. Agents Chemother. 1996, 40, 419–425. [Google Scholar] [CrossRef] [Green Version]
- Ivnitski-Steele, I.; Holmes, A.R.; Lamping, E.; Monk, B.C.; Cannon, R.D.; Sklar, L.A. Identification of Nile Red as a fluorescent substrate of the Candida albicans ABC transporters Cdr1p and Cdr2p and the MFS transporter Mdr1p. Anal. Biochem. 2009, 394, 87–91. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Makrygiannakis, D.; Revu, S.; Engström, M.; af Klint, E.; Nicholas, A.P.; Pruijn, G.J.M.; Catrina, A.I. Local administration of glucocorticoids decreases synovial citrullination in rheumatoid arthritis. Arthritis Res. Ther. 2012, 14, R20. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- LaForce, C. Use of nasal steroids in managing allergic rhinitis. J. Allergy Clin. Immunol. 1999, 103 Pt 2, S388–S394. [Google Scholar] [CrossRef]
- Cain, D.W.; Cidlowski, J.A. After 62 years of regulating immunity, dexamethasone meets COVID-19. Nat. Rev. Immunol. 2020, 20, 587–588. [Google Scholar] [CrossRef]
- Liu, N.N.; Köhler, J.R. Antagonism of fluconazole and a proton pump inhibitor against Candida albicans. Antimicrob. Agents Chemother. 2016, 60, 1145–1147. [Google Scholar] [CrossRef] [Green Version]
- Kaneko, Y.; Fukazawa, H.; Ohno, H.; Miyazaki, Y. Combinatory effect of fluconazole and FDA-approved drugs against Candida albicans. J. Infect. Chemother. 2013, 19, 1141–1145. [Google Scholar] [CrossRef]
- Urai, M.; Kaneko, Y.; Niki, M.; Inoue, M.; Tanabe, K.; Umeyama, T.; Fukazawa, H.; Ohno, H.; Miyazaki, Y. Potent drugs that attenuate anti-Candida albicans activity of fluconazole and their possible mechanisms of action. J. Infect. Chemother. 2014, 20, 612–615. [Google Scholar] [CrossRef] [PubMed]
- Monk, B.C.; Niimi, K.; Lin, S.; Knight, A.; Kardos, T.B.; Cannon, R.D.; Parshot, R.; King, A.; Lun, D.; Harding, D.R.K. Surface-active fungicidal D-peptide inhibitors of the plasma membrane proton pump that block azole resistance. Antimicrob. Agents Chemother. 2005, 49, 57–70. [Google Scholar] [CrossRef] [Green Version]
- Hayama, K.; Ishibashi, H.; Ishijima, S.A.; Niimi, K.; Tansho, S.; Ono, Y.; Monk, B.C.; Holmes, A.R.; Harding, D.R.K.; Cannon, R.D.; et al. A d-octapeptide drug efflux pump inhibitor acts synergistically with azoles in a murine oral candidiasis infection model. FEMS Microbiol. Lett. 2012, 328, 130–137. [Google Scholar] [CrossRef] [PubMed]
- Taff, H.T.; Mitchell, K.F.; Edward, J.A.; Andes, D.R. Mechanisms of Candida biofilm drug resistance. Future Microbiol. 2013, 8, 1325–1337. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nett, J.E.; Lepak, A.J.; Marchillo, K.; Anders, D.R. Time course global gene expression analysis of an in vivo Candida biofilm. J. Infect. Dis. 2009, 200, 307–313. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kelly, S.L.; Lamb, D.C.; Kelly, D.E.; Manning, N.J.; Loeffler, J.; Hebart, H.; Schumacher, U.; Einsele, H. Resistance to fluconazole and cross-resistance to amphotericin B in Candida albicans from AIDS patients caused by defective sterol Δ5,6-desaturation. FEBS Lett. 1997, 400, 80–82. [Google Scholar] [CrossRef] [Green Version]
- Watson, P.F.; Rose, M.E.; Ellis, S.W.; England, H.; Kelly, S.L. Defective sterol C5-6 desaturation and azole resistance: A new hypothesis for the mode of action of azole antifungals. Biochem. Biophys. Res. Commun. 1989, 164, 1170–1175. [Google Scholar] [CrossRef]
- Lees, N.D.; Borughton, M.C.; Sanglard, D.; Bard, M. Azole susceptibility and hyphal formation in a cytochrome P-450-deficient mutant of Candida albicans. Antimicrob. Agents Chemother. 1990, 34, 831–836. [Google Scholar] [CrossRef] [Green Version]
- Kontoyiannis, D.P.; Tarrand, J.; Prince, R.; Samonis, G.; Rolston, K.V.R. Effect of fluconazole on agar invasion by Candida albicans. J. Med. Microbiol. 2001, 50, 78–82. [Google Scholar] [CrossRef] [Green Version]
- Hitchcock, C.A.; Barrett-Bee, K.J.; Russell, N.J. The lipid composition and permeability to the triazole antifungal antibiotic ICI 153066 of serum-grown mycelial cultures of Candida albicans. J. Gen. Microbiol. 1989, 135, 1949–1955. [Google Scholar] [CrossRef] [Green Version]
- Barchiesi, F.; Di Francesco, L.F.; Scalise, G. In vitro activities of terbinafine in combination with fluconazole and itraconazole against isolates of Candida albicans with reduced susceptibility to azoles. Antimicrob. Agents Chemother. 1997, 41, 1812–1814. [Google Scholar] [CrossRef] [Green Version]
- Ghannoum, M.A.; Elewski, B. Successful treatment of fluconazole-resistant oropharyngeal candidiasis by a combination of fluconazole and terbinafine. Clin. Diagn. Lab. Immunol. 1999, 6, 921–923. [Google Scholar] [CrossRef] [Green Version]
- Kumamoto, C.A. A contact-activated kinase signals Candida albicans invasive growth and biofilms development. Proc. Natl. Acad. Sci. USA 2005, 102, 5576–5581. [Google Scholar] [CrossRef] [Green Version]
- Zhang, J.; Silao, F.G.S.; Bigol, U.G.; Bungay, A.A.C.; Nicolas, M.G.; Heitman, J.; Chen, Y.-L. Calcineurin Is Required for Pseudohyphal Growth, Virulence, and Drug Resistance in Candida lusitaniae. PLoS ONE 2012, 7, e44192. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chen, Y.-L.; Brand, A.; Morrison, E.L.; Silao, F.G.S.; Bigol, U.G.; Malbas, F.F.; Nett, J.E.; Andes, D.R.; Solis, N.V.; Filler, S.G.; et al. Calcineurin controls drug tolerance, hyphal growth, and virulence in Candida dubliniensis. Eukaryot. Cell 2011, 10, 803–819. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sanglard, D.; Ischer, F.; Marchetti, O.; Entenza, J.; Bille, J. Calcineurin A of Candida albicans: Involvement in antifungal tolerance, cell morphogenesis and virulence. Mol. Microbiol. 2003, 48, 959–976. [Google Scholar] [CrossRef] [PubMed]
- Liu, S.; Hou, Y.; Liu, W.; Lu, C.; Wang, W.; Sun, S. Components of the calcium-calcineurin signaling pathway in fungal cells and their potential as antifungal _targets. Eukaryot. Cell 2015, 14, 324–334. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lafayette, S.L.; Collins, C.; Zaas, A.K.; Schell, W.A.; Betancourt-Quiroz, M.; Leslie Gunatilaka, A.A.; Perfect, J.R.; Cowen, L.E. PKC signaling regulates drug resistance of the fungal pathogen candida albicans via circuitry comprised of mkc1, calcineurin, and hsp90. PLoS Pathog. 2010, 6, e1001069. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Singh, S.D.; Robbins, N.; Zaas, A.K.; Schell, W.A.; Perfect, J.R.; Cowen, L.E. Hsp90 governs echinocandin resistance in the pathogenic yeast Candida albicans via calcineurin. PLoS Pathog. 2009, 5, e1000532. [Google Scholar] [CrossRef] [PubMed]
- Lamoth, F.; Juvvadi, P.R.; Fortwendel, J.R.; Steinbach, W.J. Heat shock protein 90 is required for conidiation and cell wall integrity in Aspergillus fumigatus. Eukaryot. Cell 2012, 11, 1324–1332. [Google Scholar] [CrossRef] [Green Version]
- Juvvadi, P.R.; Lamoth, F.; Steinbach, W.J. Calcineurin-Mediated Regulation of Hyphal Growth, Septation, and Virulence in Aspergillus fumigatus. Mycopathologia 2014, 178, 341–348. [Google Scholar] [CrossRef] [Green Version]
- Luo, G.; Samaranayake, L.P. Candida glabrata, an emerging fungal pathogen, exhibits superior relative cell surface hydrophobicity and adhesion to denture acrylic surfaces compared with Candida albicans. Apmis 2002, 110, 601–610. [Google Scholar] [CrossRef]
- Pompilio, A.; Piccolomini, R.; Picciani, C.; D’Antonio, D.; Savini, V.; Di Bonaventura, G. Factors associated with adherence to and biofilm formation on polystyrene by Stenotrophomonas maltophilia: The role of cell surface hydrophobicity and motility. FEMS Microbiol. Lett. 2008, 287, 41–47. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Samaranayake, Y.H.; Wu, P.C.; Samaranayake, M.S. Relationship between the cell surface hydrophobicity and adherence of Candida krusei and Candida albicans to epithelial and denture acrylic surfaces. APMIS 1995, 103, 707–713. [Google Scholar] [CrossRef] [PubMed]
- Kim, J.; Guan, K.L. mTOR as a central hub of nutrient signalling and cell growth. Nat. Cell Biol. 2019, 21, 63–71. [Google Scholar] [CrossRef] [PubMed]
- Shekhar-guturja, T.; Tebung, W.A.; Mount, H.; Liu, N.; Köhler, J.R.; Whiteway, M.; Cowen, L.E. Beauvericin Potentiates Azole Activity via Inhibition of Multidrug Efflux, Blocks Candida albicans Morphogenesis, and Is Effluxed via Yor1 and Circuitry Controlled by Zcf29. Antimicrob. Agents Chemother. 2016, 60, 7468–7480. [Google Scholar] [CrossRef] [Green Version]
- Karababa, M.; Coste, A.T.; Rognon, B.; Bille, J.; Sanglard, D. Comparison of gene expression profiles of Candida albicans azole-resistant clinical isolates and laboratory strains exposed to drugs inducing multidrug transporters. Antimicrob. Agents Chemother. 2004, 48, 3064–3079. [Google Scholar] [CrossRef] [Green Version]
- Siekierka, J.J.; Sigal, N.H. FK-506 and cyclosporin A: Immunosuppressive mechanism of action and beyond. Curr. Opin. Immunol. 1992, 4, 548–552. [Google Scholar] [CrossRef]
- Lamoth, F.; Juvvadi, P.R.; Gehrke, C.; Steinbach, W.J. In vitro activity of calcineurin and heat shock protein 90 inhibitors against aspergillus fumigatus azole- and echinocandin-resistant strains. Antimicrob. Agents Chemother. 2013, 57, 1035–1039. [Google Scholar] [CrossRef] [Green Version]
- Trepel, J.; Mollapour, M.; Giaccone, G.; Neckers, L. _targeting the dynamic HSP90 complex in cancer. Nat. Rev. Cancer 2010, 10, 537–549. [Google Scholar] [CrossRef] [Green Version]
- Deorukhkar, S.C.; Saini, S.; Mathew, S. Non- albicans Candida Infection: An Emerging Threat. Interdiscip. Perspect. Infect. Dis 2014, 2014, 615958. [Google Scholar] [CrossRef] [Green Version]
- Spinillo, A.; Capuzzo, E.; Gulminetti, R.; Marone, P.; Colonna, L.; Piazzi, G. Prevalence of and risk factors for fungal vaginitis caused by non-albicans species. Am. J. Obstet. Gynecol. 1997, 176, 138–141. [Google Scholar] [CrossRef]
- Tortorano, A.M.; Prigitano, A.; Lazzarini, C.; Passera, M.; Deiana, M.L.; Cavinato, S.; De Luca, C.; Grancini, A.; Lo Cascio, G.; Ossi, C.; et al. A 1-year prospective survey of candidemia in Italy and changing epidemiology over one decade. Infection 2013, 41, 655–662. [Google Scholar] [CrossRef] [PubMed]
- Aslanyan, L.; Sanchez, D.A.; Valdebenito, S.; Eugenin, E.A.; Ramos, R.L.; Martinez, L.R. The crucial role of biofilms in Cryptococcus neoformans survival within macrophages and colonization of the central nervous system. J. Fungi 2017, 3, 10. [Google Scholar] [CrossRef] [PubMed]
- Shankar, J. An overview of toxins in Aspergillus associated with pathogenesis. Int. J. Life Sci. Biotechnol. Pharma Res. 2013, 2, 16–31. [Google Scholar]
- Eichner, R.D.; Al Salami, M.; Wood, P.R.; Müllbacher, A. The effect of gliotoxin upon macrophage function. Int. J. Immunopharmacol. 1986, 8, 789–797. [Google Scholar] [CrossRef]
- Mülbacher, A.; Eichner, R.D. Immunosuppression in vitro by a metabolite of a human pathogenic fungus. Proc. Natl. Acad. Sci. USA 1984, 81, 3835–3837. [Google Scholar] [CrossRef] [Green Version]
- Stanzani, M.; Orciuolo, E.; Lewis, R.; Kontoyiannis, D.P.; Martins, S.L.R.; St. John, L.S.; Komanduri, K.V. Aspergillus fumigatus suppresses the human cellular immune response via gliotoxin-mediated apoptosis of monocytes. Blood 2005, 105, 2258–2265. [Google Scholar] [CrossRef] [Green Version]
- Sutton, P.; Newcombe, N.R.; Waring, P.; Müllbacher, A. In vivo immunosuppressive activity of gliotoxin, a metabolite produced by human pathogenic fungi. Infect. Immun. 1994, 62, 1192–1198. [Google Scholar] [CrossRef] [Green Version]
- Amitani, R.; Taylor, G.; Elezis, E.N.; Llewellyn-Jones, C.; Mitchell, J.; Kuze, F.; Cole, P.J.; Wilson, R. Purification and characterization of factors produced by Aspergillus fumigatus which affect human ciliated respiratory epithelium. Infect. Immun. 1995, 63, 3266–3271. [Google Scholar] [CrossRef] [Green Version]
- Wang, Y.; Casadevall, A. Susceptibility of melanized and nonmelanized Cryptococcus neoformans to nitrogen- and oxygen-derived oxidants. Infect. Immun. 1994, 62, 3004–3007. [Google Scholar] [CrossRef] [Green Version]
- Jacobson, E.S.; Tinnell, S.B. Antioxidant function of fungal melanin. J. Bacteriol. 1993, 175, 7102–7104. [Google Scholar] [CrossRef] [Green Version]
- Gao, M.; Wang, H.; Zhu, L. Quercetin Assists Fluconazole to Inhibit Biofilm Formations of Fluconazole-Resistant Candida Albicans in In Vitro and in Vivo Antifungal Managements of Vulvovaginal Candidiasis. Cell. Physiol. Biochem. 2016, 40, 727–742. [Google Scholar] [CrossRef] [PubMed]
- Ahangari, F.; Farshbaf-Khalili, A.; Javadzadeh, Y.; Adibpour, M.; Sadeghzadeh Oskouei, B. Comparing the effectiveness of Salvia officinalis, clotrimazole and their combination on vulvovaginal candidiasis: A randomized, controlled clinical trial. J. Obstet. Gynaecol. Res. 2019, 45, 897–907. [Google Scholar] [CrossRef] [PubMed]
- Nouraei, S.; Amir Ali Akbari, S.; Jorjani, M.; Alavi Majd, H.; Afrakhteh, M.; Ghafoorian, A.; Tafazzoli Harandi, H. Comparison between Fluconazole with Oral Protexin Combination and Fluconazole in the Treatment of Vulvovaginal Candidiasis. ISRN Obstet. Gynecol. 2012, 2012, 375806. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Neves, N.A.; Carvalho, L.P.; Lopes, A.C.V.; Cruz, Á.; Carvalho, E.M. Successful Treatment of Refractory Recurrent Vaginal Candidiasis with Cetirizine Plus Fluconazole. J. Low. Genit. Tract Dis. 2005, 9, 167–170. [Google Scholar] [CrossRef]
- Li, S.X.; Song, Y.J.; Zhang, L.L.; Shi, J.P.; Ma, Z.L.; Guo, H.; Dong, H.Y.; Li, Y.M.; Zhang, H. An in vitro and in vivo study on the synergistic effect and mechanism of itraconazole or voriconazole alone and in combination with tetrandrine against Aspergillus fumigatus. J. Med. Microbiol. 2015, 64, 1008–1020. [Google Scholar] [CrossRef] [Green Version]
- Nakajima, R.; Kitamura, A.; Someya, K.; Tanaka, M.; Sato, K. In vitro and in vivo antifungal activities of DU-6859a, a fluoroquinolone, in combination with amphotericin B and fluconazole against pathogenic fungi. Antimicrob. Agents Chemother. 1995, 39, 1517–1521. [Google Scholar] [CrossRef] [Green Version]
- Spitzer, M.; Griffiths, E.; Blakely, K.M.; 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. [Google Scholar] [CrossRef] [Green Version]
- Sangalli-Leite, F.; Scorzoni, L.; Alves de Paula e Silva, A.C.; da Silva, J.d.F.; de Oliveira, H.C.; de Lacorte Singulani, J.; Gullo, F.P.; Moraes da Silva, R.; Regasini, L.O.; Siqueira da Silva, D.H.; et al. Synergistic effect of pedalitin and amphotericin B against Cryptococcus neoformans by in vitro and in vivo evaluation. Int. J. Antimicrob. Agents 2016, 48, 504–511. [Google Scholar] [CrossRef] [Green Version]
- Miceli, M.H.; Bernardo, S.M.; Lee, S.A. In vitro analyses of the combination of high-dose doxycycline and antifungal agents against Candida albicans biofilms. Int. J. Antimicrob. Agents 2009, 34, 326–332. [Google Scholar] [CrossRef]
- Shin, S.; Pyun, M.S. Anti-Candida effects of estragole in combination with ketoconazole or amphotericin B. Phytoher. Res. 2004, 18, 827–830. [Google Scholar] [CrossRef]
- Sadozai, S.K.; Khan, S.A.; Karim, N.; Becker, D.; Steinbrück, N.; Gier, S.; Baseer, A.; Breinig, F.; Kickelbick, G.; Schneider, M. Ketoconazole-loaded PLGA nanoparticles and their synergism against Candida albicans when combined with silver nanoparticles. J. Drug Deliv. Sci. Technol. 2020, 56, 101574. [Google Scholar] [CrossRef]
- Longhi, C.; Santos, J.P.; Morey, A.T.; Marcato, P.D.; Duran, N.; Pinge-Filho, P.; Nakazato, G.; Yamada-Ogatta, S.F.; Yamauchi, L.M. Combination of fluconazole with silver nanoparticles produced by Fusarium oxysporum improves antifungal effect against planktonic cells and biofilm of drug-resistant Candida albicans. Med. Mycol. 2016, 54, 428–432. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Khan, A.A.; Alanazi, A.M.; Jabeen, M.; Khan, S.; Malik, A. Additive potential of combination therapy against cryptococcosis employing a novel amphotericin B and fluconazole loaded dual delivery system. Eur. J. Pharm. Sci. 2018, 119, 171–178. [Google Scholar] [CrossRef] [PubMed]
- Thakur, B.; Kushawaha, S.K.; Pandit, V.; Ashawat, M.S.; Kumar, P. Formulation and Evaluation of Fixed Dose Combination Tablets of Antifungal Drugs for Candida albicans Resistant to Fluconazole. J. Drug Deliv. Ther. 2020, 10, 26–36. [Google Scholar] [CrossRef]
Antifungal Drug | Potentiators | Antibiofilm Activity Spectrum | _targets | Reference |
---|---|---|---|---|
Azoles | ||||
Ketoconazole | Bifidobacterium bifidum | C. albicans | Adhesins (Als) | [135] |
Itraconazole | Lovastatin | C. albicans | Ergosterol biosynthesis | [126] |
Itraconazole Voriconazole Posaconazole | Vorinostat | A. fumigatus A. flavus A. terreus | Drug efflux pumps Hsp90 | [114] |
Voriconazole | Geldanamycin | A. fumigatus | Hsp90 | [136] |
Miconazole | Artemisinins | C. albicans | Not identified | [92] |
Domiphen bromide | C. albicans (S & FLC-R) C. glabrata C. auris | Not identified | [118] | |
Simvastatin | C. albicans | Ergosterol biosynthesis | [120] | |
Antimycin A CCCP Sodium azide | C. albicans | Electron transport chain | [120] | |
Miconazole Voriconazole Fluconazole | 2-Adamantanamine | C. albicans (S & FLC-R) | Hyphal formation Ergosterol biosynthesis | [117] |
Fluconazole Itraconazole | Palmatine | C. albicans (S & FLC-R) C. glabrata C. krusei C. parapsilosis C. tropicalis C. guilliermondii | Drug efflux pumps | [137] |
Ketamine | C. albicans (FLC-R) | Membrane integrity ROS production Apoptosis | [138] | |
Fluconazole | Minocycline | C. neoformans (S & R) | Not identified | [115] |
Verapamil | C. albicans | Hyphal formation Adhesins (Als3) Calcium channels | [127] | |
D-penicillamine | C. albicans (S & R) | Hyphal formation Intracellular calcium homeostasis Metacaspase activation | [139] | |
Fluoxetine | C. albicans (R) | Secreted phospholipases & aspartyl proteinases | [140] | |
Ribavirin | C. albicans (S & R) | Hyphal formation Secreted phospholipases | [141] | |
Licofelone | C. albicans (S & R) | Hyphal formation Secreted phospholipases & aspartyl proteinases RAS/cAMP/PKA Pathway | [142] | |
Quercetin | C. albicans (R) | Quorum sensing Hyphal formation Secreted phospholipases & proteinases | [143] | |
Pitavastatin | C. albicans (R) C. glabrata C. auris | Drug efflux pumps | [144] | |
Dexamethasone | C. albicans (R) | Secreted phospholipases Drug efflux pumps | [145] | |
Gentamicin | C. albicans (S & R) | Secreted phospholipases Drug efflux pumps | [146] | |
Budesonide | C. albicans (R) | Secreted phospholipases Drug efflux pumps Apoptosis induction | [147] | |
Proton pump inhibitors (e.g., omeprazole, rabeprazole) | C. albicans (R) | Hyphal formation Secreted phospholipases Drug efflux pumps | [148] | |
Gypenosides | C. albicans (R) | Hyphal formation Drug efflux pumps | [149] | |
Diorcinol D | C. albicans (S & R) | Drug efflux pumps Ergosterol biosynthesis | [107] | |
Minocycline | C. albicans (S & R) | Calcium homeostasis Penetration into biofilm | [150] | |
Cyclosporine A | C. albicans | Adhesins (Als3, Hwp1) Drug efflux pumps Calcineurin Cellular surface hydrophobicity | [151,152] | |
FK506 (Tacrolimus) | C. albicans | Calcineurin | [152] | |
Geldanamycin | C. albicans | Hsp90 | [136] | |
Beauvericin | C. albicans | Hyphal formation Drug efflux pumps Hsp90 (via TORC1 kinase & CK2 kinase) | [153] | |
Polyenes | ||||
AmB | Aspirin | C. albicans C. parapsilosis | Not identified | [105] |
Toremifene citrate Drospirenone, Perhexiline maleate | C. albicans C. glabrata | Not identified | [116] | |
10 small molecule compounds | C. albicans persisters | Not identified | [124] | |
Aspartyl protease inhibitors | C. albicans | Aspartyl proteases | [131] | |
Lactoferrin | C. albicans C. glabrata | Hyphal formation | [154] | |
Deoxyribonuclease I | C. albicans A. fumigatus | Extracellular polymer matrix | [155,156] | |
Alginate lyase | A. fumigatus | Extracellular polymer matrix | [94] | |
Eugenol | C. albicans | Calcium channels ROS production Apoptosis | [157] | |
Echinocandins | ||||
Caspofungin | Toremifene citrate Drospirenone Perhexiline maleate | C. albicans C. glabrata | Not identified | [116] |
Aspartyl protease inhibitors | C. albicans | Aspartyl proteases | [131] | |
Deoxyribonuclease I | C. albicans A. fumigatus | Extracellular polymer matrix | [155,156] | |
Fluphenazine | C. glabrata (R) | Calmodulin | [158] | |
Caspofungin Micafungin | Geldanamycin | A. fumigatus | Hsp90 | [136] |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2020 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Tits, J.; Cammue, B.P.A.; Thevissen, K. Combination Therapy to Treat Fungal Biofilm-Based Infections. Int. J. Mol. Sci. 2020, 21, 8873. https://doi.org/10.3390/ijms21228873
Tits J, Cammue BPA, Thevissen K. Combination Therapy to Treat Fungal Biofilm-Based Infections. International Journal of Molecular Sciences. 2020; 21(22):8873. https://doi.org/10.3390/ijms21228873
Chicago/Turabian StyleTits, Jana, Bruno P. A. Cammue, and Karin Thevissen. 2020. "Combination Therapy to Treat Fungal Biofilm-Based Infections" International Journal of Molecular Sciences 21, no. 22: 8873. https://doi.org/10.3390/ijms21228873
APA StyleTits, J., Cammue, B. P. A., & Thevissen, K. (2020). Combination Therapy to Treat Fungal Biofilm-Based Infections. International Journal of Molecular Sciences, 21(22), 8873. https://doi.org/10.3390/ijms21228873