Preprint has been submitted for publication in journal
Preprint / Version 1

The Use of Candida auris Therapies based on Nanotechnology as Potential Novel Strategy against COVID-19 Infection

A Mini Review

Authors

  • Gerson Nakazato Universidade Estadual de Londrina
  • Wagner J Favaro Universidade Estadual de Campinas
  • Guillermo R Castro Universidad Nacional de Rosario- CONICET
  • Nelson Duran Universidade Estadual de Campinas

DOI:

https://doi.org/10.21467/preprints.337

Abstract

Candida auris, which is one of the causative agents of candidiasis, has been detected in several individuals with immune deficiency worldwide, mainly in different American countries, since 2012. C. auris infections are at risk of becoming epidemic because this species shows multi-drug resistance to several antifungal drugs available in the market; thus, since the current public health condition at global scale is threatened by the SARS-CoV-2 pandemic, C. auris infections could lead to high mortality rates. Different strategies, such as drug repurposing and the combination of antifungal drugs to other biocide molecules, were developed. However, they are time-limited strategies since drug resistance has increased due to C. auris pathologies. As an alternative, the recent development of nanotechnological devices has opened room for the efficient treatment of C. auris infections. Most specifically, the biocide effect of nanoparticles combined to/capped with antifungal drugs in different platforms seems to be an affordable technology to stop invasive C. auris infections.

Keywords:

Candidiasis, Antimicrobials, Nanoparticles, SARS-CoV-2

Downloads

Download data is not yet available.

References

Billamboz M, Fatima Z, Hameed S, Jawhara S. Promising drug candidates and new strategies for fighting against the emerging superbug Candida auris. Microorganisms 2021; 9(3): 634.

Calvo B, Melo ASA, Perozo-Mena A, et al. First report of Candida auris in America: Clinical and microbiological aspects of 18 episodes of Candidemia. J Infect 2016; 73: 369-374.

Villanueva-Lozano, H., Treviño-Rangel, R. de J., González, G.M., et al. Outbreak of Candida auris infection in a COVID-19 hospital in Mexico. Clin Microbiol Infect 2021; 27: 813-816.

Almeida Jr. JN, Francisco EC, Hagen F, et al. Emergence of Candida auris in Brazil in a COVID-19 intensive care unit. J Fungi 2021; 7: 220.

PAHO-2021. Pan American Health Organization/World Health Organization (PAHO/WHO). Candida auris outbreaks in health care services in the context of themCIVID-19 pandemia. April 6, 2021. https://www.paho.org/en/documents/epidemiological-alert-candida-auris-outbreaks-health-care-services-context-covid-19.

Rossato L. COVID-19 and Candida auris co-infection: an increasing threat. J Trop Pathol 2021; 50: 73-75.

Prestel C, Anderson E, Forsberg K, et al. Candida auris outbreak in a COVID-19 specialty care unit, Florida, July–August 2020, Morbidity mortality weekly report (US Department of Health and Human Services/Centers for Disease Control and Prevention) (MMWR) 2021; 70: 56-57

Rossato L, Colombo AL. Candida auris: What have we learned about its mechanisms of pathogenicity? Front Microbiol 2018; 9: 3081.

Larwood DJ. Nikkomycin Z-Ready to Meet the Promise? J Fungi (Basel) 2020;6 (4):261.

Bentz ML, Nunnally N, Lockhart SR, et al. Antifungal activity of nikkomycin Z against Candida auris. J Antimicrob Chemother. 2021; 76(6):1495-1497.

Chybowska AD, Childers DS, Farrer RA. Nine things genomics can tell us about Candida auris. Front Genet 2020; 11: 351.

Ahmad S, Alfouzan W. Candida auris: Epidemiology, diagnosis, pathogenesis, antifungal susceptibility, and infection control measures to combat the spread of infections in healthcare facilities. Microorganisms 2021; 9: 807.

Bidaud AL, Chowdhary A, Dannaoui E. Candida auris: An emerging drug resistant yeast – a mini-review. J Mycol Med 2018; 28: 568–573.

Bhattacharya, S., Holowka, T., Orner, et al. Gene duplication associated with increased Fluconazole tolerance in Candida auris cells of advanced generational age. Sci. Rep. 2019; 9: 5052.

Kim SH, Iyer KR., Pardeshi L, et al. Genetic analysis of Candida auris implicates Hsp90 in morphogenesis and azole tolerance and Cdr1 in azole resistance. mBio 2019; 10: e02529-18.

Kean R, Delaney C, Sherry L, et al. Transcriptome assembly and profiling of Candida auris reveals novel insights into biofilm-mediated resistance. mSphere 2018; 3: e00334-18.

Kean R, Ramage G. Combined antifungal resistance and biofilm tolerance: the global threat of Candida auris. mSphere 2019; 4: e00458-19.

Muñoz JF, Gade L, Chow NA, et al. Genomic insights into multidrug-resistance, mating and virulence in Candida auris and related emerging species. Nat Commun 2018; 9: 5346.

Muñoz JF, Welsh RM, Shea T, et al. Chromosomal rearrangements and loss of subtelomeric adhesins linked to clade-specific phenotypes in Candida auris. bioRxiv [Preprint]. 2019; doi: 10.1101/754143.

Seiler GT, Ostrosky-Zeichner L. Investigational agents for the treatment of resistant yeasts and molds. Curr Fungal Infect Rep 2021; https://doi.org/10.1007/s12281-021-00419-5.

Hager CL, Larkin EL, Long L, et al. In vitro and in vivo evaluation of the antifungal activity of APX001A/APX001 against Candida auris. Antimicrob Agents Chemother 2018; 62: e02319-17.

Berkow EL, Lockhart SR. Fluconazole resistance in Candida species: a current perspective. Infect. Drug Resist 2017; 10: 237-245.

Larkin EL, Long L, Isham N, et al. A novel 1,3-beta-d-glucan inhibitor, Ibrexafungerp (Formerly SCY-078), shows potent activity in the lower pH environment of vulvovaginitis. Antimicrob Agents Chemother 2019; 63: e02611-18.

Wiederhold NP, Lockhart SR, Najvar LK, et al. The fungal Cyp51-specific inhibitor VT-1598 demonstrates in vitro and in vivo activity against Candida auris. Antimicrob Agents Chemother 2019; 63: e02233-18.

Brand SR, Degenhardt TP, Person K, et al. A phase 2, randomized, double-blind, placebo-controlled, dose-ranging study to evaluate the efficacy and safety of orally administered VT-1161 in the treatment of recurrent vulvovaginal candidiasis. Am J Obstet Gynecol 2018; 218: 624.e1–624.e9.

Lepak AJ, Zhao M, Andes DR. Pharmacodynamic evaluation of Rezafungin (CD101) against Candida auris in the neutropenic mouse invasive candidiasis model. Antimicrob Agents Chemother 2018; 62:e01572-18.

Angulo D. Open-Label Study to Evaluate the Efficacy and Safety of Oral Ibrexafungerp (SCY-078) in Patients With Candidiasis Caused by Candida Auris (CARES). https://clinicaltrials.gov/ct2/show/NCT03363841.

Nishikawa H, Fukuda Y, Mitsuyama J. et al. In vitro and in vivo antifungal activities of T-2307, a novel arylamidine, against Cryptococcus gattii: An emerging fungal pathogen. J Antimicrob Chemother 2017; 72: 1709-1713.

Murray A, Cass L, Ito K, et al. PC945, a novel inhaled antifungal agent, for the treatment of respiratory fungal infections. J Fungi 2020; 6: 373.

Singh R, Kaur M, Chakrabarti A et al. Biofilm formation by Candida auris isolated from colonising sites and candidemia cases. Mycoses 2019; 62: 706-709.

Leach MD, Farrer RA, Tan K, et al. Hsf1 and Hsp90 orchestrate temperature-dependent global transcriptional remodelling and chromatin architecture in Candida albicans. Nat Commun 2016; 7:11704.

Chakrabarti A, Sood P. On the emergence, spread and resistance of Candida auris: Host, pathogen and environmental tipping points. J Med Microbiol 2021; 70: 001318.

OMS, 2020. Antimicrobial resistance. https://www.who.int/news-room/fact-sheets/detail/antimicrobial-resistance. (Accessed on July 2, 2021)

Misra SK, Pathak K. Supercritical fluid technology for solubilization of poorly water-soluble drugs via micro- and nanosized particle generation. ADMET and DMPK, 2020; 8: 355-374.

Emig D, Ivliev A, Pustovalova O, et al. Drug target prediction and repositioning using an integrated network-based approach. PLoS One. 2013; 8: e60618.

Aghaei Gharehbolagh S, Izadi A, Talebi M, et al. New weapons to fight a new enemy: A systematic review of drug combinations against the drug-resistant fungus Candida auris. Mycoses. 2021, In press. doi: 10.1111/myc.13277.

Shamshina JL, Barber PS, Rogers RD. Ionic liquids in drug delivery. Expert Opin Drug Deliv 2013; 10: 1367-1381.

Vandana KR, Prasanna Raju Y, Harini Chowdary V, et al. An overview on in situ micronization technique - An emerging novel concept in advanced drug delivery. Saudi Pharm J 2014; 22: 283-289.

Xue H, Li J, Xie H, Wang Y. Review of drug repositioning approaches and resources. Int J Biol Sci 2018; 14: 1232-1244.

Ahmad A, Wei Y, Syed F, et al. Amphotericin B-conjugated biogenic silver nanoparticles as an innovative strategy for fungal infections. Microb Pathogen 2016; 99: 271–281.

Amaral AC, Saavedra PHV, Oliveira Souza AC, et al., Miconazole loaded chitosan-based nanoparticles for local treatment of vulvovaginal candidiasis fungal infections. Colloids Surf B Biointerfaces. 2019; 174: 409-415.

Araújo J, Martín-Pastor M, Pérez L, et al. Development of anacardic acid-loaded zein nanoparticles: Physical chemical characterization, stability, and antimicrobial improvement. J Mol Liq 2021; 332: 115808.

Albertini B, Passerini N, Di Sabatino M, et al. Polymer-lipid based mucoadhesive microspheres prepared by spray-congealing for the vaginal delivery of econazole nitrate, Eur J Pharm Sci 2009; 36(4-5): 591–601.

Monteiro DR, Gorup LF, Silva S, et al. Silver colloidal nanoparticles: antifungal effect against adhered cells and biofilms of Candida albicans and Candida glabrata. Biofouling. 2011; 27(7):711-9.

Lara HH, Ixtepan-Turrent L, Yacaman MJ, et al. Inhibition of Candida auris biofilm formation on medical and environmental surfaces by silver nanoparticles. ACS Appl Mater Interf 2020; 12: 19.

Vazquez-Muñoz R, Lopez FD, Lopez-Ribot JL. Silver Nanoantibiotics display strong antifungal activity against the emergent multidrug-resistant yeast Candida auris under both planktonic and biofilm growing conditions. Front Microbiol 2020a; 11:1673.

Gangadoo S, Elbourne A, Medvedev AE. et al. Facile route of fabricating long-term microbicidal silver nanoparticle clusters against Shiga toxin-producing Escherichia coli O157:H7 and Candida auris. Coatings 2020; 10: 28.

Vazquez-Muñoz R, Lopez FD, Lopez-Ribot JL. Bismuth nanoantibiotics display anticandidal activity and disrupt the biofilm and cell morphology of the emergent pathogenic yeast Candida auris. Antibiotics 2020b; 9: 461.

Bonilla JJA, Honorato L, de Oliveira DFC, et al. Silver chitosan nanocomposites as a potential treatment for superficial candidiasis. Med Mycol 2021; myab028. https://doi.org/10.1093/mmy/myab028

Gupta A, Briffa SM, Swingler S, et al. Synthesis of silver nanoparticles using curcumin-cyclodextrins loaded into bacterial cellulose based hydrogels for wound dressing applications. Biomacromolecules 2020; 21:1802-1811.

Sherin P, Kuriakose S. Synthesis of Superparamagnetic iron oxide nanoparticles stabilized by biocompatible supramolecular ?-cyclodextrin for biomedical applications. Mater Today Proc 2019; 11:1030-1035.

Kamli MR, Srivastava V, Hajrah NH, et al. Phytogenic fabrication of Ag-Fe bimetallic nanoparticles for cell cycle arrest and apoptosis signaling pathways in Candida auris by generating oxidative stress. Antioxidants (Basel, Switzerland) 2021a; 10(2): 182.

Kamli MR, Srivastava V, Hajrah NH, et al. Facile bio-fabrication of Ag-Cu-Co trimetallic nanoparticles and its fungicidal activity against Candida auris. J Fungi 2021b; 7: 62.

Cleare LG, Li KL, Abuzeid WM, et al. NO Candida auris: Nitric oxide in nanotherapeutics to combat emerging fungal pathogen Candida auris. J Fungi 2020; 6: 85.

Barreto TL, Rossato L, de Freitas ALD, et al. Miltefosine as an alternative strategy in the treatment of the emerging fungus Candida auris. Inter J Antimicrob Agents 2020; 56: 106049.

Spadari CC, de Bastiani FWMDS, Lopes LB, Ishida K. Alginate nanoparticles as non-toxic delivery system for miltefosine in the treatment of candidiasis and cryptococcosis. Int J Nanomedicine 2019; 14: 5187-5199.

Hamdy R, Fayed B, Hamoda AM, et al. Essential oil-based design and development of novel anti-candida azoles formulation. Molecules 2020; 25, 1463.

Zhang L, Li X, Zhu S, et al. Dermal targeting delivery of terbinafine hydrochloride using novel multi-ethosomes: A new approach to fungal infection treatment. Coatings 2020; 10: 304.

Silva C de B, Guterres SS, Weisheimer V, Schapoval EE. Antifungal activity of the lemongrass oil and citral against Candida spp. Braz J Infect Dis 2008; 12(1):63-66.

Ahmad A, Khan A, Akhtar F, et al. Fungicidal activity of thymol and carvacrol by disrupting ergosterol biosynthesis and membrane integrity against Candida. Eur J Clin Microbiol Infect Dis. 2011; 30(1): 41-50.

Shreaz S, Wani WA, Behbehani JM, et al. Cinnamaldehyde and its derivatives, a novel class of antifungal agents. Fitoterapia. 2016; 112:116-131.

Miranda-Cadena K, Dias M, Costa-Barbosa A, et al. Development and characterization of monoolein-based liposomes of carvacrol, cinnamaldehyde, citral, or thymol with Anti-Candida Activities. Antimicrob Agents Chemother 2021; 65(4):e01628-1620.

Hornby JM, Jensen EC, Lisec AD, et al. Quorum sensing in the dimorphic fungus Candida albicans is mediated by farnesol. Appl Environ Microbiol 2001; 67(7): 2982-2992.

Langford ML, Atkin AL, Nickerson KW. Cellular interactions of farnesol, a quorum-sensing molecule produced by Candida albicans. Future Microbiol 2009; 4: 1353-1362.

Nagy F, Vitális E, Jakab Á, et al. In vitro and in vivo effect of exogenous farnesol exposure against Candida auris. Front Microbiol. 2020a;11: 957.

Nagy F, Tóth Z, Daróczi L, et al. Farnesol increases the activity of echinocandins against Candida auris biofilms. Med Mycol. 2020b; 58(3): 404-407.

Srivastava V, Ahmad A. Abrogation of pathogenic attributes in drug resistant Candida auris strains by farnesol. PLoS One 2020; 15(5): e0233102.

Bezerra CF, de Alencar Júnior JG, de Lima Honorato R, et al. Antifungal activity of farnesol incorporated in liposomes and associated with fluconazole. Chem Phys Lipids 2020; 233:104987.

Cavassin FB, Baú-Carneiro JL, Vilas-Boas RR., et al. Sixty years of Amphotericin B: An overview of the main antifungal agent used to treat invasive fungal infections. Infect dis ther 2021; 10: 115–147.

Wang X, Mohammad IS, Fan L, et al. Delivery strategies of amphotericin B for invasive fungal infections, Acta Pharm Sin B 2021; In press. https://doi.org/10.1016/j.apsb.2021.04.010

Hamill, RJ. Amphotericin B formulations: a comparative review of efficacy and toxicity. Drugs 2013; 73(9): 919–934.

Herrada J, Gamal A, Long L, et al. In vitro and in vivo antifungal activity of AmBisome compared to conventional amphotericin B and fluconazole against Candida auris. Antimicrob Agents Chemother 2021; 65(6):e00306-21.

Rodriguez YJ, Quejada LF, Villamil JC, et al. Development of amphotericin B micellar formulations based on copolymers of poly (ethylene glycol) and poly(?-caprolactone) conjugated with retinol. Pharmaceutics 2020; 12: 196.

Chatzimoschou A, Giampani A, Meis JF, et al. Activities of nine antifungal agents against Candida auris biofilms. Mycoses 2020; 64: 381-384.

Griffith N, Larry Danziger L. Candida auris urinary tract infections and possible treatment. Antibiotics 2020; 9: 898.

Alves D, Vaz AT, Grainha T, et al. Design of an antifungal surface embedding liposomal amphotericin b through a mussel adhesive-inspired coating strategy. Front Chem 2019; 7: 431.

Gudz KY, Antipina LY, Permyakova ES, et al., Ag-doped and antibiotic-loaded hexagonal boron nitride nanoparticles as promising carriers to fight different pathogens. ACS Appl Mater Interfaces 2021; 13(20): 23452-23468.

Tutaj K, Szlazak R, Szalapata K, et al. Amphotericin B-silver hybrid nanoparticles: synthesis, properties and antifungal activity. Nanomed-nanotechnol 2016; 12: 1095–1103.

Jafari M, Abolmaali SS, Tamaddon AM, et al. Nanotechnology approaches for delivery and targeting of Amphotericin B in fungal and parasitic diseases. Nanomedicine (London, England) 2021; 16: 857–877.

Gudz KY, Permyakova ES, Matveev AT, et al. Pristine and antibiotic-loaded nanosheets/nanoneedles-based boron nitride films as a promising platform to suppress bacterial and fungal Infections. ACS Appl Mater Interfaces 2020; 12(38):42485-42498.

Dennis EK, Kim JH, Parkin S, et al. Distorted Gold(I)-Phosphine Complexes as Antifungal Agents. J Med Chem 2020; 63(5): 2455-2469.

Domingues Bianchin M, Borowicz SM, da Rosa Monte Machado G, et al. Lipid core nanoparticles as a broad strategy to reverse fluconazole resistance in multiple Candida species. Colloids Surf B Biointerfaces. 2019; 175:523-529.

Sadeghi Ghadi Z, Dinarvand R, Asemi N, et al. Preparation, characterization and in vivo evaluation of novel hyaluronan containing niosomes tailored by Box-Behnken design to co-encapsulate curcumin and quercetin. Eur J Pharm Sci 2019; 130: 234-246.

Sadeghi-Ghadi Z, Vaezi A, Ahangarkani F, et al. Potent in vitro activity of curcumin and quercetin co-encapsulated in nanovesicles without hyaluronan against Aspergillus and Candida isolates. J Mycol Med 2020; 30(4): 101014.

Hashemi MM, Rovig J, Holden BS, et al. Ceragenins are active against drug-resistant Candida auris clinical isolates in planktonic and biofilm forms. J Antimicrob Chemother 2018; 73(6):1537-1545.

Eldesouky HE, Lanman NA, Hazbun TR, et al. Aprepitant, an antiemetic agent, interferes with metal ion homeostasis of Candida auris and displays potent synergistic interactions with azole drugs. Virulence 2020; 11(1): 1466-1481.

Kim HR, Eom YB. Antifungal and anti-biofilm effects of 6-shogaol against Candida auris. J Appl Microbiol. 2021; 130(4): 1142-1153.

Downloads

Posted

2021-08-29