451
|
Powell LC, Adams JYM, Quoraishi S, Py C, Oger A, Gazze SA, Francis LW, von Ruhland C, Owens D, Rye PD, Hill KE, Pritchard MF, Thomas DW. Alginate oligosaccharides enhance the antifungal activity of nystatin against candidal biofilms. Front Cell Infect Microbiol 2023; 13:1122340. [PMID: 36798083 PMCID: PMC9927220 DOI: 10.3389/fcimb.2023.1122340] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 01/11/2023] [Indexed: 02/04/2023] Open
Abstract
Background The increasing prevalence of invasive fungal infections in immuno-compromised patients is a considerable cause of morbidity and mortality. With the rapid emergence of antifungal resistance and an inadequate pipeline of new therapies, novel treatment strategies are now urgently required. Methods The antifungal activity of the alginate oligosaccharide OligoG in conjunction with nystatin was tested against a range of Candida spp. (C. albicans, C. glabrata, C. parapsilosis, C. auris, C. tropicalis and C. dubliniensis), in both planktonic and biofilm assays, to determine its potential clinical utility to enhance the treatment of candidal infections. The effect of OligoG (0-6%) ± nystatin on Candida spp. was examined in minimum inhibitory concentration (MIC) and growth curve assays. Antifungal effects of OligoG and nystatin treatment on biofilm formation and disruption were characterized using confocal laser scanning microscopy (CLSM), scanning electron microscopy (SEM) and ATP cellular viability assays. Effects on the cell membrane were determined using permeability assays and transmission electron microscopy (TEM). Results MIC and growth curve assays demonstrated the synergistic effects of OligoG (0-6%) with nystatin, resulting in an up to 32-fold reduction in MIC, and a significant reduction in the growth of C. parapsilosis and C. auris (minimum significant difference = 0.2 and 0.12 respectively). CLSM and SEM imaging demonstrated that the combination treatment of OligoG (4%) with nystatin (1 µg/ml) resulted in significant inhibition of candidal biofilm formation on glass and clinical grade silicone surfaces (p < 0.001), with increased cell death (p < 0.0001). The ATP biofilm disruption assay demonstrated a significant reduction in cell viability with OligoG (4%) alone and the combined OligoG/nystatin (MIC value) treatment (p < 0.04) for all Candida strains tested. TEM studies revealed the combined OligoG/nystatin treatment induced structural reorganization of the Candida cell membrane, with increased permeability when compared to the untreated control (p < 0.001). Conclusions Antimicrobial synergy between OligoG and nystatin against Candida spp. highlights the potential utility of this combination therapy in the prevention and topical treatment of candidal biofilm infections, to overcome the inherent tolerance of biofilm structures to antifungal agents.
Collapse
Affiliation(s)
- Lydia C. Powell
- Advanced Therapies Group, Cardiff University School of Dentistry, Cardiff, United Kingdom
- Microbiology and Infectious Disease group, Swansea University Medical School, Swansea, United Kingdom
- *Correspondence: Lydia C. Powell,
| | - Jennifer Y. M. Adams
- Advanced Therapies Group, Cardiff University School of Dentistry, Cardiff, United Kingdom
| | - Sadik Quoraishi
- Otolaryngology Department, New Cross Hospital, Wolverhampton, United Kingdom
| | - Charlène Py
- Advanced Therapies Group, Cardiff University School of Dentistry, Cardiff, United Kingdom
- School of Engineering, University of Angers, Angers, France
| | - Anaϊs Oger
- Advanced Therapies Group, Cardiff University School of Dentistry, Cardiff, United Kingdom
- School of Engineering, University of Angers, Angers, France
| | - Salvatore A. Gazze
- Centre for Nanohealth, Swansea University Medical School, Swansea, United Kingdom
| | - Lewis W. Francis
- Centre for Nanohealth, Swansea University Medical School, Swansea, United Kingdom
| | - Christopher von Ruhland
- Central Biotechnology Services, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - David Owens
- Head and Neck Directorate, University Hospital of Wales, Cardiff, United Kingdom
| | | | - Katja E. Hill
- Advanced Therapies Group, Cardiff University School of Dentistry, Cardiff, United Kingdom
| | - Manon F. Pritchard
- Advanced Therapies Group, Cardiff University School of Dentistry, Cardiff, United Kingdom
| | - David W. Thomas
- Advanced Therapies Group, Cardiff University School of Dentistry, Cardiff, United Kingdom
| |
Collapse
|
452
|
Rawson TM, Antcliffe DB, Wilson RC, Abdolrasouli A, Moore LSP. Management of Bacterial and Fungal Infections in the ICU: Diagnosis, Treatment, and Prevention Recommendations. Infect Drug Resist 2023; 16:2709-2726. [PMID: 37168515 PMCID: PMC10166098 DOI: 10.2147/idr.s390946] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 04/22/2023] [Indexed: 05/13/2023] Open
Abstract
Bacterial and fungal infections are common issues for patients in the intensive care unit (ICU). Large, multinational point prevalence surveys have identified that up to 50% of ICU patients have a diagnosis of bacterial or fungal infection at any one time. Infection in the ICU is associated with its own challenges. Causative organisms often harbour intrinsic and acquired mechanisms of drug-resistance, making empiric and targeted antimicrobial selection challenging. Infection in the ICU is associated with worse clinical outcomes for patients. We review the epidemiology of bacterial and fungal infection in the ICU. We discuss risk factors for acquisition, approaches to diagnosis and management, and common strategies for the prevention of infection.
Collapse
Affiliation(s)
- Timothy M Rawson
- Health Protection Research Unit in Healthcare Associated Infections and Antimicrobial Resistance, Hammersmith Hospital, London, UK
- Centre for Antimicrobial Optimisation, Imperial College London, Imperial College London, London, UK
- David Price Evan’s Group in Infectious Diseases and Global Health, Department of Pharmacology and Therapeutics, University of Liverpool, Liverpool, UK
- Correspondence: Timothy M Rawson, Health Protection Research Unit in Healthcare Associated Infections & Antimicrobial Resistance, Hammersmith Hospital, Du Cane Road, London, W12 0NN, United Kingdom, Email
| | - David B Antcliffe
- Centre for Antimicrobial Optimisation, Imperial College London, Imperial College London, London, UK
- Division Anaesthesia, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Imperial College London, London, UK
| | - Richard C Wilson
- Health Protection Research Unit in Healthcare Associated Infections and Antimicrobial Resistance, Hammersmith Hospital, London, UK
- Centre for Antimicrobial Optimisation, Imperial College London, Imperial College London, London, UK
- David Price Evan’s Group in Infectious Diseases and Global Health, Department of Pharmacology and Therapeutics, University of Liverpool, Liverpool, UK
| | | | - Luke S P Moore
- Health Protection Research Unit in Healthcare Associated Infections and Antimicrobial Resistance, Hammersmith Hospital, London, UK
- Chelsea & Westminster NHS Foundation Trust, London, UK
- North West London Pathology, Imperial College Healthcare NHS Trust, London, UK
| |
Collapse
|
453
|
Xue B, Geng X, Cui H, Chen H, Wu Z, Chen H, Li H, Zhou Z, Zhao M, Tan C, Li J. Size engineering of 2D MOF nanosheets for enhanced photodynamic antimicrobial therapy. CHINESE CHEM LETT 2023. [DOI: 10.1016/j.cclet.2023.108140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
454
|
Yu J, He C, Wang T, Zhang G, Li J, Zhang J, Kang W, Xu Y, Zhao Y. Rapid automated antifungal susceptibility testing system for yeasts based on growth characteristics. Front Cell Infect Microbiol 2023; 13:1153544. [PMID: 37201120 PMCID: PMC10185846 DOI: 10.3389/fcimb.2023.1153544] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Accepted: 04/14/2023] [Indexed: 05/20/2023] Open
Abstract
Fungal pathogens are a major threat to public health, as they are becoming increasingly common and resistant to treatment, with only four classes of antifungal medicines currently available and few candidates in the clinical development pipeline. Most fungal pathogens lack rapid and sensitive diagnostic techniques, and those that exist are not widely available or affordable. In this study, we introduce a novel automated antifungal susceptibility testing system, Droplet 48, which detects the fluorescence of microdilution wells in real time and fits growth characteristics using fluorescence intensity over time. We concluded that all reportable ranges of Droplet 48 were appropriate for clinical fungal isolates in China. Reproducibility within ±2 two-fold dilutions was 100%. Considering the Sensititre YeastOne Colorimetric Broth method as a comparator method, eight antifungal agents (fluconazole, itraconazole, voriconazole, caspofungin, micafungin, anidulafungin, amphotericin B, and 5-flucytosine) showed an essential agreement of >90%, except for posaconazole (86.62%). Category agreement of four antifungal agents (fluconazole, caspofungin, micafungin, and anidulafungin) was >90%, except for voriconazole (87.93% agreement). Two Candida albicans isolates and anidulafungin showed a major discrepancy (MD) (2.60%), and no other MD or very MD agents were found. Therefore, Droplet 48 can be considered as an optional method that is more automated and can obtain results and interpretations faster than previous methods. However, the optimization of the detection performance of posaconazole and voriconazole and promotion of Droplet 48 in clinical microbiology laboratories still require further research involving more clinical isolates in the future.
Collapse
Affiliation(s)
- Jinhan Yu
- Department of Clinical Laboratory, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
- Graduate School, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Beijing Key Laboratory for Mechanisms Research and Precision Diagnosis of Invasive Fungal Diseases, Beijing, China
| | - Chun He
- Department of Clinical Laboratory, Peking University School and Hospital of Stomatology, Beijing, China
| | - Tong Wang
- Department of Clinical Laboratory, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
- Beijing Key Laboratory for Mechanisms Research and Precision Diagnosis of Invasive Fungal Diseases, Beijing, China
| | - Ge Zhang
- Department of Clinical Laboratory, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
- Beijing Key Laboratory for Mechanisms Research and Precision Diagnosis of Invasive Fungal Diseases, Beijing, China
| | - Jin Li
- Department of Clinical Laboratory, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
- Beijing Key Laboratory for Mechanisms Research and Precision Diagnosis of Invasive Fungal Diseases, Beijing, China
| | - Jingjia Zhang
- Department of Clinical Laboratory, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
- Beijing Key Laboratory for Mechanisms Research and Precision Diagnosis of Invasive Fungal Diseases, Beijing, China
| | - Wei Kang
- Department of Clinical Laboratory, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
- Beijing Key Laboratory for Mechanisms Research and Precision Diagnosis of Invasive Fungal Diseases, Beijing, China
| | - Yingchun Xu
- Department of Clinical Laboratory, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
- Beijing Key Laboratory for Mechanisms Research and Precision Diagnosis of Invasive Fungal Diseases, Beijing, China
- *Correspondence: Ying Zhao, ; Yingchun Xu,
| | - Ying Zhao
- Department of Clinical Laboratory, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
- Beijing Key Laboratory for Mechanisms Research and Precision Diagnosis of Invasive Fungal Diseases, Beijing, China
- *Correspondence: Ying Zhao, ; Yingchun Xu,
| |
Collapse
|
455
|
Takano T, Kudo H, Eguchi S, Matsumoto A, Oka K, Yamasaki Y, Takahashi M, Koshikawa T, Takemura H, Yamagishi Y, Mikamo H, Kunishima H. Inhibitory effects of vaginal Lactobacilli on C andida albicans growth, hyphal formation, biofilm development, and epithelial cell adhesion. Front Cell Infect Microbiol 2023; 13:1113401. [PMID: 37201113 PMCID: PMC10188118 DOI: 10.3389/fcimb.2023.1113401] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 04/19/2023] [Indexed: 05/20/2023] Open
Abstract
Introduction Antifungal agents are not always efficient in resolving vulvovaginal candidiasis (VVC), a common genital infection caused by the overgrowth of Candida spp., including Candida albicans, or in preventing recurrent infections. Although lactobacilli (which are dominant microorganisms constituting healthy human vaginal microbiota) are important barriers against VVC, the Lactobacillus metabolite concentration needed to suppress VVC is unknown. Methods We quantitatively evaluated Lactobacillus metabolite concentrations to determine their effect on Candida spp., including 27 vaginal strains of Lactobacillus crispatus, L. jensenii, L. gasseri, Lacticaseibacillus rhamnosus, and Limosilactobacillus vaginalis, with inhibitory abilities against biofilms of C. albicans clinical isolates. Results Lactobacillus culture supernatants suppressed viable fungi by approximately 24%-92% relative to preformed C. albicans biofilms; however, their suppression differed among strains and not species. A moderate negative correlation was found between Lactobacillus lactate production and biofilm formation, but no correlation was observed between hydrogen peroxide production and biofilm formation. Both lactate and hydrogen peroxide were required to suppress C. albicans planktonic cell growth. Lactobacillus strains that significantly inhibited biofilm formation in culture supernatant also inhibited C. albicans adhesion to epithelial cells in an actual live bacterial adhesion competition test. Discussion Healthy human microflora and their metabolites may play important roles in the development of new antifungal agent against C. albicans-induced VVC.
Collapse
Affiliation(s)
- Tomonori Takano
- Department of Infectious Diseases, St. Marianna University School of Medicine, Kawasaki-shi, Kanagawa, Japan
| | - Hayami Kudo
- Research Department, R&D Division, Miyarisan Pharmaceutical Co., Ltd., Saitama-shi, Saitama, Japan
| | - Shuhei Eguchi
- Research Department, R&D Division, Miyarisan Pharmaceutical Co., Ltd., Saitama-shi, Saitama, Japan
| | - Asami Matsumoto
- Research Department, R&D Division, Miyarisan Pharmaceutical Co., Ltd., Saitama-shi, Saitama, Japan
| | - Kentaro Oka
- Research Department, R&D Division, Miyarisan Pharmaceutical Co., Ltd., Saitama-shi, Saitama, Japan
| | - Yukitaka Yamasaki
- Department of Infectious Diseases, St. Marianna University School of Medicine, Kawasaki-shi, Kanagawa, Japan
| | - Motomichi Takahashi
- Research Department, R&D Division, Miyarisan Pharmaceutical Co., Ltd., Saitama-shi, Saitama, Japan
| | - Takuro Koshikawa
- Department of Microbiology, St. Marianna University School of Medicine, Kawasaki-shi, Japan
| | - Hiromu Takemura
- Department of Microbiology, St. Marianna University School of Medicine, Kawasaki-shi, Japan
| | - Yuka Yamagishi
- Department of Clinical Infectious Diseases, Aichi Medical University, Nagakute, Aichi, Japan
- Department of Clinical Infectious Diseases, Kochi Medical School, Nankoku-shi, Kochi, Japan
| | - Hiroshige Mikamo
- Department of Clinical Infectious Diseases, Aichi Medical University, Nagakute, Aichi, Japan
| | - Hiroyuki Kunishima
- Department of Infectious Diseases, St. Marianna University School of Medicine, Kawasaki-shi, Kanagawa, Japan
- *Correspondence: Hiroyuki Kunishima,
| |
Collapse
|
456
|
Mossion A, Ourliac-Garnier I, Wielgosz-Collin G. Fungal Sterol Analyses by Gas Chromatography-Mass Spectrometry Using Different Derivatives. Methods Mol Biol 2023; 2704:143-156. [PMID: 37642842 DOI: 10.1007/978-1-0716-3385-4_8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
Sterols are the main components of the fungal membrane. Their study can be used to describe the chemical biodiversity among the strains and species or to work on antifungal treatment. Those molecules can be analyzed by gas chromatography coupled with mass spectrometry (GC-MS) as free molecules or after derivation as acetate or trimethylsilyl ether (TMSi). Here we describe how to extract sterols from fungal biomass according to its physiological form and the culture conditions (liquid and solid media). The different methodologies that can be used to obtain free sterols, acetate, and/or TMSi derivatives are presented. Identification keys using the fragmentation at 70 eV are also described.
Collapse
Affiliation(s)
- Aurélie Mossion
- Nantes Université, Institut des Substances et Organismes de la Mer, ISOMer, Nantes, France.
| | - Isabelle Ourliac-Garnier
- Nantes Université, Cibles et Médicaments des Infections et de l'Immunité, IICiMed, Nantes, France
| | | |
Collapse
|
457
|
Sun LL, Li H, Yan TH, Cao YB, Jiang YY, Yang F. Aneuploidy enables cross-tolerance to unrelated antifungal drugs in Candida parapsilosis. Front Microbiol 2023; 14:1137083. [PMID: 37113223 PMCID: PMC10126355 DOI: 10.3389/fmicb.2023.1137083] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 03/17/2023] [Indexed: 04/29/2023] Open
Abstract
Candida parapsilosis is an emerging major human fungal pathogen. Echinocandins are first-line antifungal drugs for the treatment of invasive Candida infections. In clinical isolates, tolerance to echinocandins in Candida species is mostly due to point mutations of FKS genes, which encode the target protein of echinocandins. However, here, we found chromosome 5 trisomy was the major mechanism of adaptation to the echinocandin drug caspofungin, and FKS mutations were rare events. Chromosome 5 trisomy conferred tolerance to echinocandin drugs caspofungin and micafungin and cross-tolerance to 5-flucytosine, another class of antifungal drugs. The inherent instability of aneuploidy caused unstable drug tolerance. Tolerance to echinocandins might be due to increased copy number and expression of CHS7, which encodes chitin synthase. Although copy number of chitinase genes CHT3 and CHT4 was also increased to the trisomic level, the expression was buffered to the disomic level. Tolerance to 5-flucytosine might be due to the decreased expression of FUR1. Therefore, the pleiotropic effect of aneuploidy on antifungal tolerance was due to the simultaneous regulation of genes on the aneuploid chromosome and genes on euploid chromosomes. In summary, aneuploidy provides a rapid and reversible mechanism of drug tolerance and cross-tolerance in C. parapsilosis.
Collapse
Affiliation(s)
- Liu-liu Sun
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
- Department of Physiology and Pharmacology, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Hao Li
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
- Department of Physiology and Pharmacology, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Tian-hua Yan
- Department of Physiology and Pharmacology, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Yong-bing Cao
- Department of Vascular Diseases, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yuan-ying Jiang
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
- Yuan-ying Jiang
| | - Feng Yang
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
- *Correspondence: Feng Yang
| |
Collapse
|
458
|
Meng F, Liu X, Li C, Peng X, Wang Q, Xu Q, Sui J, Zhao G, Lin J. Hinokitiol inhibits Aspergillus fumigatus by interfering with the cell membrane and cell wall. Front Microbiol 2023; 14:1132042. [PMID: 37113218 PMCID: PMC10128913 DOI: 10.3389/fmicb.2023.1132042] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Accepted: 03/20/2023] [Indexed: 04/29/2023] Open
Abstract
Hinokitiol (β-thujaplicin) is an important component of the essential oil extracted from Chamaecyparis obtuse, which prevents the decay and decomposition of temple and shrine buildings in Japan. Hinokiol has been shown to have a detrimental effect on various fungi such as Candida albicans and saprophytic fungi. However how hinokitiol works against Aspergillus fumigatus (A. fumigatus) has not been claimed. This study aims to investigate the adverse effects of hinokitiol on the disruption of the cell wall and cell membrane of A. fumigatus and to explore possible potential mechanisms or pathways. According to our results, hinokitiol negatively altered mycelium morphology, growth density, and cell plasma composition content. When incubated with human corneal epithelial cells (HCECs), hinokitiol saw a safe effect with concentrations below 12 μg/ml. Hinokitiol was shown to increase the cell membrane's permeability by decreasing the cell membrane's ergosterol content. The integrity of the cell wall was disrupted, as well as a significant increase in chitin degradation and chitinase activity. As determined by RNA-seq results, subsequent analysis, and qRT-PCR, altered transcript levels of cell walls and cell membranes-related genes (such as eglC) illustrated how hinokitiol affected the genetic profile of A. fumigatus. With this study, we recommend hinokitiol as an effective anti-A. fumigatus agent by reducing the amounts of key components in the cell wall and membrane by preventing production and accelerating breakdown.
Collapse
|
459
|
Salmanton-García J, Hoenigl M, Gangneux JP, Segal E, Alastruey-Izquierdo A, Arikan Akdagli S, Lagrou K, Özenci V, Vena A, Cornely OA. The current state of laboratory mycology and access to antifungal treatment in Europe: a European Confederation of Medical Mycology survey. THE LANCET. MICROBE 2023; 4:e47-e56. [PMID: 36463916 DOI: 10.1016/s2666-5247(22)00261-0] [Citation(s) in RCA: 56] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 08/16/2022] [Accepted: 09/02/2022] [Indexed: 12/04/2022]
Abstract
Access to the appropriate tools is crucial for early diagnosis and clinical management of invasive fungal infections. This Review aims to describe the invasive fungal infection diagnostic capacity of Europe to better understand the status and the most pressing aspects that need improvement. To our knowledge, this is the first time that the mycological diagnostic capability and access to antifungal treatments of institutions has been evaluated at a pan-European level. Between Nov 1, 2021, and Jan 31, 2022, 388 institutions in Europe self-assessed their invasive fungal infection management capability. Of the 388 participating institutions from 45 countries, 383 (99%) had access to cultures, 375 (97%) to microscopy, 363 (94%) to antigen-detection assays, 329 (85%) to molecular tests (mostly PCR), and 324 (84%) to antibody tests for diagnosis and management. With the exception of microscopy, there were considerable differences in access to techniques among countries according to their gross domestic product. At least one triazole was available in 363 (94%) of the institutions, one echinocandin in 346 (89%), and liposomal amphotericin B in 301 (78%), with country gross domestic product-based differences. Differences were also observed in the access to therapeutic drug monitoring. Although Europe is well prepared to manage invasive fungal infections, some institutions do not have access to certain diagnostic tools and antifungal drugs, despite most being considered essential by WHO. These limitations need to be overcome to ensure that all patients receive the best diagnostic and therapeutic management.
Collapse
Affiliation(s)
- Jon Salmanton-García
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany; Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD) and Excellence Center for Medical Mycology, University of Cologne, Cologne, Germany; Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany.
| | - Martin Hoenigl
- Division of Infectious Diseases, ECMM Center of Excellence for Medical Mycology, Medical University of Graz, Graz, Austria; Division of Infectious Diseases and Global Public Health, Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Jean-Pierre Gangneux
- CHU de Rennes, INSERM, Institut de Recherche en Santé, Environnement et Travail, (UMR_S 1085), University of Rennes, Rennes, France
| | - Esther Segal
- Department of Clinical Microbiology and Immunology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ana Alastruey-Izquierdo
- Mycology Reference Laboratory, Spanish National Centre for Microbiology, Instituto de Salud Carlos III, Madrid, Spain
| | - Sevtap Arikan Akdagli
- Hacettepe University Faculty of Medicine, Department of Medical Microbiology, Ankara, Türkiye
| | - Katrien Lagrou
- Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium; Department of Laboratory Medicine, Center of Excellence for Medical Mycology, and National Reference Center for Mycosis, UZ Leuven, Leuven, Belgium
| | - Volkan Özenci
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden; Department of Clinical Microbiology, Karolinska University Hospital, Stockholm, Sweden
| | - Antonio Vena
- Department of Health Sciences, University of Genoa, Genoa, Italy; Infectious Diseases Unit, San Martino Policlinico Hospital, IRCCS for Oncology and Neuroscience, Genoa, Italy
| | - Oliver A Cornely
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany; Clinical Trials Centre Cologne, University of Cologne, Cologne, Germany; Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD) and Excellence Center for Medical Mycology, University of Cologne, Cologne, Germany; Centre for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany; Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| |
Collapse
|
460
|
Dilek Altintop M, Cantürk Z, Özdemir A. A series of 2-pyrazolines endowed with potent anticandidal activity. MAKEDONSKO FARMACEVTSKI BILTEN 2022. [DOI: 10.33320/maced.pharm.bull.2022.68.03.202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Affiliation(s)
- Mehlika Dilek Altintop
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Anadolu University, 26470 Eskişehir, Turkey
| | - Zerrin Cantürk
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Anadolu University, 26470 Eskişehir, Turkey
| | - Ahmet Özdemir
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Anadolu University, 26470 Eskişehir, Turkey
| |
Collapse
|
461
|
Uroro EO, Bright R, Hayles A, Vasilev K. Lipase-Responsive Amphotericin B Loaded PCL Nanoparticles for Antifungal Therapies. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 13:155. [PMID: 36616065 PMCID: PMC9823996 DOI: 10.3390/nano13010155] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 12/22/2022] [Accepted: 12/27/2022] [Indexed: 06/17/2023]
Abstract
Amphotericin B is an antifungal drug used for the treatment of invasive fungal infections. However, its clinical use is limited due to its serious side effects, such as renal and cardiovascular toxicity. Furthermore, amphotericin B is administered in high doses due to its poor water solubility. Hence, it is necessary to develop an on-demand release strategy for the delivery of amphotericin B to reduce cytotoxicity. The present report describes a novel encapsulation of amphotericin B into lipase-sensitive polycaprolactone to form a nanocomposite. Nanocomposites were produced by the oil-in-water method and their physicochemical properties such as size, hydrodynamic diameter, drug loading, and zeta potential were determined. The in vitro release of amphotericin B was characterized in the presence and absence of lipase. The antifungal activity of the nanocomposites was verified against lipase-secreting Candida albicans, and cytotoxicity was tested against primary human dermal fibroblasts. In the absence of lipase, the release of amphotericin B from the nanocomposites was minimal. However, in the presence of lipase, an enzyme that is abundant at infection sites, a fungicidal concentration of amphotericin B was released from the nanocomposites. The antifungal activity of the nanocomposites showed an enhanced effect against the lipase-secreting fungus, Candida albicans, in comparison to the free drug at the same concentration. Furthermore, nanoencapsulation significantly reduced amphotericin B-related cytotoxicity compared to the free drug. The synthesized nanocomposites can serve as a potent carrier for the responsive delivery of amphotericin B in antifungal applications.
Collapse
Affiliation(s)
| | - Richard Bright
- College of Medicine and Public Health, Flinders University, Bedford Park, SA 5042, Australia
| | - Andrew Hayles
- College of Medicine and Public Health, Flinders University, Bedford Park, SA 5042, Australia
| | - Krasimir Vasilev
- UniSA STEM, University of South Australia, Mawson Lakes, SA 5095, Australia
- College of Medicine and Public Health, Flinders University, Bedford Park, SA 5042, Australia
| |
Collapse
|
462
|
Khan FA, Yaqoob S, Ali S, Tanveer N, Wang Y, Ashraf S, Hasan KA, Khalifa SAM, Shou Q, Ul-Haq Z, Jiang ZH, El-Seedi HR. Designing Functionally Substituted Pyridine-Carbohydrazides for Potent Antibacterial and Devouring Antifungal Effect on Multidrug Resistant (MDR) Strains. MOLECULES (BASEL, SWITZERLAND) 2022; 28:molecules28010212. [PMID: 36615406 PMCID: PMC9822510 DOI: 10.3390/molecules28010212] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 12/21/2022] [Accepted: 12/21/2022] [Indexed: 12/28/2022]
Abstract
The emergence of multidrug-resistant (MDR) pathogens and the gradual depletion of available antibiotics have exacerbated the need for novel antimicrobial agents with minimal toxicity. Herein, we report functionally substituted pyridine carbohydrazide with remarkable antimicrobial effect on multi-drug resistant strains. In the series, compound 6 had potent activity against four MDR strains of Candida spp., with minimum inhibitory concentration (MIC) values being in the range of 16-24 µg/mL and percentage inhibition up to 92.57%, which was exceptional when compared to broad-spectrum antifungal drug fluconazole (MIC = 20 µg/mL, 81.88% inhibition). Substitution of the octyl chain in 6 with a shorter butyl chain resulted in a significant anti-bacterial effect of 4 against Pseudomonas aeruginosa (ATCC 27853), the MIC value being 2-fold superior to the standard combination of ampicillin/cloxacillin. Time-kill kinetics assays were used to discern the efficacy and pharmacodynamics of the potent compounds. Further, hemolysis tests confirmed that both compounds had better safety profiles than the standard drugs. Besides, molecular docking simulations were used to further explore their mode of interaction with target proteins. Overall results suggest that these compounds have the potential to become promising antimicrobial drugs against MDR strains.
Collapse
Affiliation(s)
- Farooq-Ahmad Khan
- Third World Center for Science and Technology, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
- H.E.J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
- Correspondence: (F.-A.K.); (K.A.H.); (H.R.E.-S.)
| | - Sana Yaqoob
- Third World Center for Science and Technology, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
- H.E.J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Shujaat Ali
- Third World Center for Science and Technology, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
- H.E.J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Nimra Tanveer
- Third World Center for Science and Technology, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
- H.E.J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Yan Wang
- H.E.J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Sajda Ashraf
- H.E.J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Khwaja Ali Hasan
- Molecular and Structural Biology Research Laboratory, Department of Biochemistry, University of Karachi, Karachi 75270, Pakistan
- Correspondence: (F.-A.K.); (K.A.H.); (H.R.E.-S.)
| | - Shaden A. M. Khalifa
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, S-106 91 Stockholm, Sweden
| | - Qiyang Shou
- Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Zaheer Ul-Haq
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Zi-Hua Jiang
- Department of Chemistry, Lakehead University, 955 Oliver Road, Thunder Bay, Ontario P7B 5E1, Canada
| | - Hesham R. El-Seedi
- International Research Center for Food Nutrition and Safety, Jiangsu University, Zhenjiang 212013, China
- Pharmacognosy Group, Department of Pharmaceutical Biosciences, BMC, Uppsala University, Box 591, SE-751 24 Uppsala, Sweden
- International Joint Research Laboratory of Intelligent Agriculture and Agri-Products Processing (Jiangsu University), Jiangsu Education Department, Nanjing 210024, China
- Correspondence: (F.-A.K.); (K.A.H.); (H.R.E.-S.)
| |
Collapse
|
463
|
Jean SS, Yang HJ, Hsieh PC, Huang YT, Ko WC, Hsueh PR. In Vitro Susceptibilities of Worldwide Isolates of Intrapulmonary Aspergillus Species and Important Candida Species in Sterile Body Sites against Important Antifungals: Data from the Antimicrobial Testing Leadership and Surveillance Program, 2017-2020. Microbiol Spectr 2022; 10:e0296522. [PMID: 36314941 PMCID: PMC9769544 DOI: 10.1128/spectrum.02965-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 09/27/2022] [Indexed: 11/07/2022] Open
Abstract
To understand the changes of resistance in clinically commonly encountered fungi, we used the Antimicrobial Testing Leadership and Surveillance (ATLAS) database to explore in vitro antifungal susceptibilities against clinically important isolates of Aspergillus and Candida species (collected from intrapulmonary and sterile body areas, respectively). We applied the CLSI antifungal 2020 and the EUCAST antifungal 2020 guidelines. From 2017 to 2020, isolates of intrapulmonary Aspergillus fumigatus (n = 660), Aspergillus niger (n = 107), Aspergillus flavus (n = 96), Aspergillus terreus (n = 40), and Aspergillus nidulans species complex (n = 26) and sterile site-originated isolates of Candida albicans (n = 1,810), Candida glabrata (n = 894), Candida krusei (n = 120), Candida dubliniensis (n = 107), Candida lusitaniae (n = 82), Candida guilliermondii (n = 28), and Candida auris (n = 7) were enrolled in this study. Using the EUCAST 2020 breakpoints, it was demonstrated that amphotericin B and posaconazole displayed poor in vitro susceptibility rates against A. fumigatus isolates (<50% and 18.9%, respectively). In contrast, isavuconazole and itraconazole showed high in vitro potency against most Aspergillus isolates (>92%). Most intrapulmonary Aspergillus isolates exhibited MICs of ≤0.06 μg/mL to anidulafungin. Furthermore, intrapulmonary A. fumigatus isolates collected from Italy and the United Kingdom exhibited lower in vitro susceptibility to isavuconazole (72.2% and 69%, respectively) than those in the remaining ATLAS participant countries (>85%). Higher isavuconazole MIC90s against C. auris and C. guilliermondii (1 and 4 μg/mL, respectively) were observed compared to the other five Candida species. Despite the aforementioned MICs and susceptibilities against fungi, research needs to consider the pharmacokinetic (PK) profiles, pharmacodynamic (PD) parameters, and clinical treatment experience with antifungals against specific Aspergillus species. IMPORTANCE In addition to monitoring the antifungal susceptibilities of clinically important fungi, reviewing the PK/PD indices and the clinical therapy experience of antifungals under evaluation are important to guide an appropriate antifungal prescription. The efficacies of liposomal amphotericin B complex and anidulafungin for the treatment of pulmonary aspergillosis caused by different Aspergillus species need to be periodically evaluated in the future.
Collapse
Affiliation(s)
- Shio-Shin Jean
- Department of Internal Medicine, Min-Sheng General Hospital, Taoyuan, Taiwan
- Department of Pharmacy, College of Pharmacy and Health Care, Tajen University, Pingtung, Taiwan
- Department of Critical Care Medicine, Min-Sheng General Hospital, Taoyuan, Taiwan
| | - Hung-Jen Yang
- Department of Family Medicine, Min-Sheng General Hospital, Taoyuan, Taiwan
| | - Po-Chuen Hsieh
- Department of Pharmacy, College of Pharmacy and Health Care, Tajen University, Pingtung, Taiwan
| | - Yu-Tsung Huang
- Department of Laboratory Medicine, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, Taiwan
- Department of Internal Medicine, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Wen-Chien Ko
- Department of Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Po-Ren Hsueh
- Department of Laboratory Medicine, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, Taiwan
- Department of Laboratory Medicine, China Medical University Hospital, China Medical University, Taichung, Taiwan
- School of Medicine, China Medical University, Taichung, Taiwan
- Ph.D Program for Aging, School of Medicine, China Medical University, Taichung, Taiwan
- Department of Internal Medicine, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, Taiwan
- Department of Internal Medicine, China Medical University Hospital, China Medical University, Taichung, Taiwan
| |
Collapse
|
464
|
Malathi S, Manikandan D, Nishanthi R, Jagan EG, Riyaz SUM, Palani P, Simal‐Gandara J. Silver Nanoparticles, Synthesized using
Hyptis suaveolens
(L) Poit and their Antifungal Activity against
Candida
spp. ChemistrySelect 2022. [DOI: 10.1002/slct.202203050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Selvaraj Malathi
- Centre for Advanced Studies in Botany University of Madras, Guindy Campus Chennai India
- Department of Biotechnology Sri Sankara Arts and Science College (Autonomous) Enathur Kanchipuram
| | - Dhayalan Manikandan
- Small Molecules and Drug Discovery group, Chengdu Anticancer Biosciecne, Tianfu International Biotown Chengdu 610000 China
| | - Ramasami Nishanthi
- Department of Biotechnology College of Science and Humanities, SRMIST Kattankulathur 603 203 Tamilnadu India
| | - Enthai Ganeshan Jagan
- Department of Biotechnology Sri Sankara Arts and Science College (Autonomous) Enathur Kanchipuram
- Department of Molecular Microbiology School of Biotechnology Madurai Kamaraj University Madurai -625021 India
| | - Savaas Umar Mohammed Riyaz
- PG & Research Department of Biotechnology Islamiah College (Autonomous) Vaniyambadi 635752 Tamilnadu India
| | - Perumal Palani
- Centre for Advanced Studies in Botany University of Madras, Guindy Campus Chennai India
| | - Jesus Simal‐Gandara
- Universidade de Vigo Nutrition and Bromatology Group Analytical Chemistry and Food Science Department Faculty of Science E-32004 Ourense Spain
| |
Collapse
|
465
|
Griffiths JS, Orr SJ, Morton CO, Loeffler J, White PL. The Use of Host Biomarkers for the Management of Invasive Fungal Disease. J Fungi (Basel) 2022; 8:jof8121307. [PMID: 36547640 PMCID: PMC9784708 DOI: 10.3390/jof8121307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 12/03/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022] Open
Abstract
Invasive fungal disease (IFD) causes severe morbidity and mortality, and the number of IFD cases is increasing. Exposure to opportunistic fungal pathogens is inevitable, but not all patients with underlying diseases increasing susceptibility to IFD, develop it. IFD diagnosis currently uses fungal biomarkers and clinical risk/presentation to stratify high-risk patients and classifies them into possible, probable, and proven IFD. However, the fungal species responsible for IFD are highly diverse and present numerous diagnostic challenges, which culminates in the empirical anti-fungal treatment of patients at risk of IFD. Recent studies have focussed on host-derived biomarkers that may mediate IFD risk and can be used to predict, and even identify IFD. The identification of novel host genetic variants, host gene expression changes, and host protein expression (cytokines and chemokines) associated with increased risk of IFD has enhanced our understanding of why only some patients at risk of IFD actually develop disease. Furthermore, these host biomarkers when incorporated into predictive models alongside conventional diagnostic techniques enhance predictive and diagnostic results. Once validated in larger studies, host biomarkers associated with IFD may optimize the clinical management of populations at risk of IFD. This review will summarise the latest developments in the identification of host biomarkers for IFD, their use in predictive modelling and their potential application/usefulness for informing clinical decisions.
Collapse
Affiliation(s)
- James S. Griffiths
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral and Craniofacial Sciences, King’s College London, London WC2R 2LS, UK
| | - Selinda J. Orr
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Science, Queen’s University Belfast, Belfast BT9 7BL, UK
| | | | - Juergen Loeffler
- Department of Internal Medicine II, University Hospital of Würzburg, 97070 Würzburg, Germany
| | - P. Lewis White
- Public Health Wales, Microbiology Cardiff, University Hospital of Wales, Heath Park, Cardiff CF14 4XW, UK
- Correspondence:
| |
Collapse
|
466
|
Firacative C, Zuluaga-Puerto N, Guevara J. Cryptococcus neoformans Causing Meningoencephalitis in Adults and a Child from Lima, Peru: Genotypic Diversity and Antifungal Susceptibility. J Fungi (Basel) 2022; 8:jof8121306. [PMID: 36547639 PMCID: PMC9781953 DOI: 10.3390/jof8121306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 12/13/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022] Open
Abstract
Cryptococcosis, caused predominantly by Cryptococcus neoformans, is a potentially fatal, opportunistic infection that commonly affects the central nervous system of immunocompromised patients. Globally, this mycosis is responsible for almost 20% of AIDS-related deaths, and in countries like Peru, its incidence remains high, mostly due to the annual increase in new cases of HIV infection. This study aimed to establish the genotypic diversity and antifungal susceptibility of C. neoformans isolates causing meningoencephalitis in 25 adults and a 9-year-old girl with HIV and other risk factors from Lima, Peru. To identify the genotype of the isolates, multilocus sequence typing was applied, and to establish the susceptibility of the isolates to six antifungals, a YeastOne® broth microdilution was used. From the isolates, 19 were identified as molecular type VNI, and seven as VNII, grouped in eight and three sequence types, respectively, which shows that the studied population was highly diverse. Most isolates were susceptible to all antifungals tested. However, VNI isolates were less susceptible to fluconazole, itraconazole and voriconazole than VNII isolates (p < 0.05). This study contributes data on the molecular epidemiology and the antifungal susceptibility profile of the most common etiological agent of cryptococcosis, highlighting a pediatric case, something which is rare among cryptococcal infection.
Collapse
Affiliation(s)
- Carolina Firacative
- Studies in Translational Microbiology and Emerging Diseases (MICROS) Research Group, School of Medicine and Health Sciences, Universidad de Rosario, Bogota 111221, Colombia
- Correspondence:
| | | | - José Guevara
- Facultad de Medicina “San Fernando”, Universidad Nacional Mayor de San Marcos, Lima 15081, Peru
| |
Collapse
|
467
|
Study of Prescription-Indication of Outpatient Systemic Anti-Fungals in a Colombian Population. A Cross-Sectional Study. Antibiotics (Basel) 2022; 11:antibiotics11121805. [PMID: 36551462 PMCID: PMC9774786 DOI: 10.3390/antibiotics11121805] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 11/25/2022] [Accepted: 12/02/2022] [Indexed: 12/14/2022] Open
Abstract
The inappropriate use of antifungals is associated with greater antimicrobial resistance, costs, adverse events, and worse clinical outcomes. The aim of this study was to determine prescription patterns and approved and unapproved indications for systemic antifungals in a group of patients in Colombia. This was a cross-sectional study on indications for the use of systemic antifungals in outpatients from a drug dispensing database of approximately 9.2 million people affiliated with the Colombian Health System. Sociodemographic, pharmacological, and clinical variables were considered. Descriptive, bivariate, and multivariate analyses were performed. A total of 74,603 patients with antifungal prescriptions were identified; they had a median age of 36.0 years (interquartile range: 22.0−53.0 years), and 67.3% of patients were women. Fluconazole (66.5%) was the most prescribed antifungal for indications such as vaginitis, vulvitis, and vulvovaginitis (35.0%). A total of 29.3% of the prescriptions were used in unapproved indications. A total of 96.3% of ketoconazole users used the medication in unapproved indications. Men (OR: 1.91; CI95%: 1.79−2.04), <18 years of age (OR: 1.20; CI95%: 1.11−1.31), from the Caribbean region (OR: 1.26; CI95%: 1.18−1.34), with chronic obstructive pulmonary disease (OR: 1.80; CI95%: 1.27−2.54), prescriptions made by a general practitioner (OR: 1.17; CI95%: 1.04−1.31), receiving comedications (OR: 1.58; CI95%: 1.48−1.69), and the concomitant use of other antimicrobials (OR: 1.77; CI95%: 1.66−1.88) were associated with a higher probability that the antifungal was used for unapproved indications; deep mycosis (OR: 0.49; CI95%: 0.41−0.58), prescribing fluconazole (OR: 0.06; CI95%: 0.06−0.06), and having diabetes mellitus (OR: 0.33; CI95%: 0.29−0.37), cancer (OR: 0.13; CI95%: 0.11−0.16), or HIV (OR: 0.07; CI95%: 0.04−0.09) reduced this risk. Systemic antifungals were mostly used for the management of superficial mycoses, especially at the gynecological level. In addition, more than a quarter of patients received these medications in unapproved indications, and there was broad inappropriate use of ketoconazole.
Collapse
|
468
|
Antifungal and Anti-Inflammatory Activities of PS1-2 Peptide against Fluconazole-Resistant Candida albicans. Antibiotics (Basel) 2022; 11:antibiotics11121779. [PMID: 36551434 PMCID: PMC9774446 DOI: 10.3390/antibiotics11121779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 12/06/2022] [Accepted: 12/07/2022] [Indexed: 12/13/2022] Open
Abstract
Clinically, fungal pneumonia rarely occurs in adults, and invasive fungal infections can cause substantial morbidity, and mortality due to sepsis and septic shock. In the present study, we have designed peptides that exhibit potent antifungal activities against fluconazole-resistant Candida albicans in physiological monovalent, and divalent ionic buffers, with minimal fungicidal concentrations ranging from 16 to 32 µM. None of these tested peptides resulted in the development of drug resistance similar to fluconazole. Among them, the PS1-2 peptide did not induce stimulation of macrophages by C. albicans, and it exerted antifungal and anti-inflammatory effects against C. albicans-induced intratracheal infection, in an acute lung injury mouse model. PS1-2 is likely a novel therapeutic agent for the control, and prevention of drug-resistant C. albicans infection, and our findings may be useful for designing antimicrobial peptides to combat fungal infection.
Collapse
|
469
|
Zhai P, Ma Y, Du W, Lu L. The metal chaperone protein MtmA plays important roles in antifungal drug susceptibility in Aspergillus fumigatus. Front Microbiol 2022; 13:1062282. [DOI: 10.3389/fmicb.2022.1062282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 11/21/2022] [Indexed: 12/04/2022] Open
Abstract
Drug-resistant fungal infections are emerging as an important clinical problem. In general, antifungal resistance results from increased target expression or mutations within the target protein sequence. However, the molecular mechanisms of non-drug target mutations of antifungal resistance in fungal pathogens remain to be explored. Previous studies indicated that the metal chaperone protein Mtm1 is required for mitochondrial Sod2 activation and responses to oxidative stress in yeast and in the fungal pathogen Aspergillus fumigatus, but there is no report of MtmA-related antifungal resistance. In this study, we found that repressed expression of MtmA (only 10% expression) using a conditional promoter resulted in significantly enhanced itraconazole resistance, which was not the result of highly expressed drug targets Erg11A and Erg11B. Furthermore, we demonstrated that repressed expression of MtmA results in upregulation of a series of multidrug resistance-associated transport genes, which may cause multidrug resistance. Further mechanistic studies revealed that inhibition of MtmA expression led to abnormal activation of the calcium signaling system and prompted persistent nucleation of the calcium signaling transcription factor CrzA. Our findings suggest that the metal chaperone protein MtmA is able to negatively regulate fungal resistance via affecting calcium signaling pathway.
Collapse
|
470
|
Parums DV. Editorial: The World Health Organization (WHO) Fungal Priority Pathogens List in Response to Emerging Fungal Pathogens During the COVID-19 Pandemic. Med Sci Monit 2022; 28:e939088. [PMID: 36453055 PMCID: PMC9724454 DOI: 10.12659/msm.939088] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 11/29/2022] [Indexed: 07/29/2023] Open
Abstract
The COVID-19 pandemic, climate change, increased resistance to antifungal drugs, and an increased number of immunocompromised patients have driven a recent global surge in pathogenic fungal infections, including aspergillosis, candidiasis, and mucormycosis. On 25 October 2022, the World Health Organization (WHO) released a list of 19 fungal priority pathogens identified as having the greatest threat to public health. The WHO Fungal Priority Pathogens List represents the first global response to identify and prioritize fungal pathogens and their impact on global public health and to consider the unmet research and development needs. The WHO has grouped the priority fungal pathogens into those of critical, high, and medium priority. This Editorial aims to highlight the importance of identifying and prioritizing fungal pathogens and identifying emerging fungal pathogens and the global factors driving changing patterns of infection.
Collapse
Affiliation(s)
- Dinah V Parums
- Science Editor, Medical Science Monitor, International Scientific Information, Inc., Melville, NY, USA
| |
Collapse
|
471
|
Verweij PE, Arendrup MC, Alastruey-Izquierdo A, Gold JAW, Lockhart SR, Chiller T, White PL. Dual use of antifungals in medicine and agriculture: How do we help prevent resistance developing in human pathogens? Drug Resist Updat 2022; 65:100885. [PMID: 36283187 PMCID: PMC10693676 DOI: 10.1016/j.drup.2022.100885] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 10/17/2022] [Accepted: 10/18/2022] [Indexed: 11/27/2022]
Abstract
Azole resistance in Aspergillus fumigatus is a One Health resistance threat, where azole fungicide exposure compromises the efficacy of medical azoles. The use of the recently authorized fungicide ipflufenoquin, which shares its mode-of-action with a new antifungal olorofim, underscores the need for risk assessment for dual use of antifungals.
Collapse
Affiliation(s)
- Paul E Verweij
- Department of Medical Microbiology and Radboudumc-CWZ Center of Expertise for Mycology, Radboud University Medical Center, Nijmegen, the Netherlands; Centre for Infectious Diseases Research, Diagnostics and Laboratory Surveillance, National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands.
| | - Maiken C Arendrup
- Unit for Mycology, Statens Serum Insitut, Copenhagen, Denmark; Department of Medical Microbiology, University Hospital Rigshospitalet, Copenhagen, Denmark; Department of Clinical Medicine, Copenhagen University, Copenhagen, Denmark
| | | | - Jeremy A W Gold
- Mycotic Diseases Branch, Centers for Disease Control and Prevention, Atlanta, USA
| | - Shawn R Lockhart
- Mycotic Diseases Branch, Centers for Disease Control and Prevention, Atlanta, USA
| | - Tom Chiller
- Mycotic Diseases Branch, Centers for Disease Control and Prevention, Atlanta, USA
| | - P Lewis White
- Public Health Wales Mycology Reference Laboratory, Cardiff, United Kingdom
| |
Collapse
|
472
|
Costa J, Sepúlveda M, Gallardo V, Cayún Y, Santander C, Ruíz A, Reyes M, Santos C, Cornejo P, Lima N, Santos C. Antifungal Potential of Capsaicinoids and Capsinoids from the Capsicum Genus for the Safeguarding of Agrifood Production: Advantages and Limitations for Environmental Health. Microorganisms 2022; 10:microorganisms10122387. [PMID: 36557640 PMCID: PMC9788535 DOI: 10.3390/microorganisms10122387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 11/21/2022] [Accepted: 11/29/2022] [Indexed: 12/05/2022] Open
Abstract
Opportunistic pathogenic fungi arise in agricultural crops as well as in surrounding human daily life. The recent increase in antifungal-resistant strains has created the need for new effective antifungals, particularly those based on plant secondary metabolites, such as capsaicinoids and capsinoids produced by Capsicum species. The use of such natural compounds is well-aligned with the One Health approach, which tries to find an equilibrium among people, animals, and the environment. Considering this, the main objective of the present work is to review the antifungal potential of capsaicinoids and capsinoids, and to evaluate the environmental and health impacts of biofungicides based on these compounds. Overall, capsaicinoids and their analogues can be used to control pathogenic fungi growth in plant crops, as eco-friendly alternatives to pest management, and assist in the conservation and long-term storage of agrifood products. Their application in different stages of the agricultural and food production chains improves food safety, nutritional value, and overcomes antimicrobial resistance, with a lower associated risk to humans, animals, and the environment than that of synthetic fungicides and pesticides. Nevertheless, research on the effect of these compounds on bee-like beneficial insects and the development of new preservatives and packaging materials is still necessary.
Collapse
Affiliation(s)
- Jéssica Costa
- Departamento de Biologia, Instituto de Ciências Biológicas-ICB, Universidade Federal do Amazonas, Av. Rodrigo Otávio Jordão Ramos 3000, Bloco 01, Manaus 69077-000, AM, Brazil
| | - Marcela Sepúlveda
- Department of Chemical Science and Natural Resources, Universidad de La Frontera, Temuco 4811-230, Chile
| | - Víctor Gallardo
- Programa de Doctorado en Ciencias de Recursos Naturales, Universidad de La Frontera, Temuco 4811-230, Chile
| | - Yasna Cayún
- Department of Chemical Science and Natural Resources, Universidad de La Frontera, Temuco 4811-230, Chile
| | - Christian Santander
- Department of Chemical Science and Natural Resources, Universidad de La Frontera, Temuco 4811-230, Chile
- Environmental Engineering and Biotechnology Group, Faculty of Environmental Science and EULA-Chile Center, Universidad de Concepción, Concepción 4070-411, Chile
| | - Antonieta Ruíz
- Department of Chemical Science and Natural Resources, Universidad de La Frontera, Temuco 4811-230, Chile
| | - Marjorie Reyes
- Department of Chemical Science and Natural Resources, Universidad de La Frontera, Temuco 4811-230, Chile
| | - Carla Santos
- CEB-Centre of Biological Engineering, Micoteca da Universidade do Minho (MUM), University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal
- LABBELS (Associate Laboratory, Braga/Guimarães), University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal
| | - Pablo Cornejo
- Escuela de Agronomía, Facultad de Ciencias Agronómicas y de los Alimentos, Pontificia Universidad Católica de Valparaíso, Quillota 2260-000, Chile
| | - Nelson Lima
- CEB-Centre of Biological Engineering, Micoteca da Universidade do Minho (MUM), University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal
- LABBELS (Associate Laboratory, Braga/Guimarães), University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal
| | - Cledir Santos
- Department of Chemical Science and Natural Resources, Universidad de La Frontera, Temuco 4811-230, Chile
- Correspondence: ; Tel.: +56-452-596-726
| |
Collapse
|
473
|
Nosratabadi M, Espahbodi A, Hedayati MT, Shokohi T, Badali H, Saeedi M, Moazeni M, Aghili SR, Javidnia J, Faeli L, Khojasteh S, Roohi B, Abbasi K, Abastabar M, Haghani I. In Vitro Combination of Terbinafine with Ketoconazole Against Aspergillus Species with Terbinafine High MIC Values Isolated From Otomycosis. Mycopathologia 2022; 188:119-127. [PMID: 36449121 DOI: 10.1007/s11046-022-00698-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 11/20/2022] [Indexed: 12/03/2022]
Abstract
Otomycosis is a common mycotic infection of the external auditory canal, and Aspergillus species are one of the most frequent causative agents worldwide. The limited antifungal arsenal, the high toxicity and side effects of antifungal agents, and the growing resistance to the currently available antifungals underscore the need for new therapeutic strategies. The present study aimed to evaluate the combined in vitro efficacy of terbinafine and ketoconazole against Aspergillus species with terbinafine high MIC values isolated from patients with otomycosis.84 Aspergillus species with high MIC values to terbinafine (≥ 4 µg/ml), consisting of A. flavus, A. tubingensis, A. niger, and A. terreus, were included in this study. The checkerboard microdilution method evaluated the in vitro interactions using the CLSI reference technique. Synergistic effects were observed for 66.67% (56/84) of all isolates (FICI ranging from 0.19 to 0.5). However, the interactions of terbinafine and ketoconazole exhibited indifference in 33.33% (28/84) of the isolates, and no antagonism was observed for any combination. The interaction of terbinafine and ketoconazole showed synergistic activity against Aspergillus species with high MIC values, suggesting that this is an alternative and promising approach for treating otomycosis.
Collapse
Affiliation(s)
- Mohsen Nosratabadi
- Department of Medical Mycology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Mazandaran Province, Iran
| | - Amirreza Espahbodi
- Student Research Committee, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mohammad Taghi Hedayati
- Department of Medical Mycology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Mazandaran Province, Iran
- Invasive Fungi Research Center, Communicable Diseases Institute, Mazandaran University of Medical Sciences, Sari, Iran
| | - Tahereh Shokohi
- Department of Medical Mycology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Mazandaran Province, Iran
- Invasive Fungi Research Center, Communicable Diseases Institute, Mazandaran University of Medical Sciences, Sari, Iran
| | - Hamid Badali
- Department of Medical Mycology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Mazandaran Province, Iran
- Invasive Fungi Research Center, Communicable Diseases Institute, Mazandaran University of Medical Sciences, Sari, Iran
- Department of Molecular Microbiology & Immunology, South Texas Center for Emerging Infectious Diseases, The University of Texas at San Antonio, San Antonio, TX, USA
| | - Majid Saeedi
- Department of Pharmaceutics, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Maryam Moazeni
- Department of Medical Mycology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Mazandaran Province, Iran
- Invasive Fungi Research Center, Communicable Diseases Institute, Mazandaran University of Medical Sciences, Sari, Iran
| | - Seyed Reza Aghili
- Department of Medical Mycology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Mazandaran Province, Iran
- Invasive Fungi Research Center, Communicable Diseases Institute, Mazandaran University of Medical Sciences, Sari, Iran
| | - Javad Javidnia
- Department of Medical Mycology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Mazandaran Province, Iran
- Student Research Committee, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Leila Faeli
- Department of Medical Mycology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Mazandaran Province, Iran
| | - Shaghayegh Khojasteh
- Department of Medical Mycology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Mazandaran Province, Iran
| | - Behrad Roohi
- Department of Medical Mycology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Mazandaran Province, Iran
| | - Kiana Abbasi
- Department of Microbiology, Zanjan Branch, Islamic Azad University, Zanjan, Iran
| | - Mahdi Abastabar
- Department of Medical Mycology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Mazandaran Province, Iran.
- Invasive Fungi Research Center, Communicable Diseases Institute, Mazandaran University of Medical Sciences, Sari, Iran.
| | - Iman Haghani
- Department of Medical Mycology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Mazandaran Province, Iran.
- Invasive Fungi Research Center, Communicable Diseases Institute, Mazandaran University of Medical Sciences, Sari, Iran.
| |
Collapse
|
474
|
Veerapandian R, Abdul Azees PA, Viswanathan T, Amaechi BT, Vediyappan G. Editorial: Developing therapeutics for antimicrobial resistant pathogens. Front Cell Infect Microbiol 2022; 12:1083501. [PMID: 36506021 PMCID: PMC9731126 DOI: 10.3389/fcimb.2022.1083501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Accepted: 11/14/2022] [Indexed: 11/25/2022] Open
Affiliation(s)
- Raja Veerapandian
- Center of Emphasis in Infectious Diseases, Department of Molecular and Translational Medicine, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, TX, United States,*Correspondence: Raja Veerapandian, ; Bennett Tochukwu Amaechi, ; Govindsamy Vediyappan,
| | - Parveez Ahamed Abdul Azees
- Department of Comprehensive Dentistry, School of Dentistry, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Thiruselvam Viswanathan
- Department of Cellular Biology and Pharmacology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, United States
| | - Bennett Tochukwu Amaechi
- Department of Comprehensive Dentistry, School of Dentistry, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States,*Correspondence: Raja Veerapandian, ; Bennett Tochukwu Amaechi, ; Govindsamy Vediyappan,
| | - Govindsamy Vediyappan
- Division of Biology, Kansas State University, Manhattan, KS, United States,*Correspondence: Raja Veerapandian, ; Bennett Tochukwu Amaechi, ; Govindsamy Vediyappan,
| |
Collapse
|
475
|
Martín-Cruz L, Angelina A, Baydemir I, Bulut Ö, Subiza JL, Netea MG, Domínguez-Andrés J, Palomares O. Candida albicans V132 induces trained immunity and enhances the responses triggered by the polybacterial vaccine MV140 for genitourinary tract infections. Front Immunol 2022; 13:1066383. [PMID: 36505433 PMCID: PMC9729253 DOI: 10.3389/fimmu.2022.1066383] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 11/09/2022] [Indexed: 11/25/2022] Open
Abstract
Introduction Recurrent urinary tract infections (RUTIs) and recurrent vulvovaginal candidiasis (RVVCs) represent major healthcare problems all over the world. Antibiotics and antifungals are widely used for such infectious diseases, which is linked with microbial resistances and microbiota deleterious effects. The development of novel approaches for genitourinary tract infections (GUTIs) such as trained immunity-based vaccines (TIbV) is therefore highly required. MV140 is a sublingual whole-cell heat-inactivated polybacterial preparation with demonstrated clinical efficacy for RUTIs. The sublingual heat-inactivated Candida albicans vaccine V132 has been developed for RVVCs. We previously showed that the combination of MV140 and V132 promotes potent Th1/Th17 and regulatory T-cell responses against antigens contained in the formulation and unrelated antigens. The specific contribution of each preparation to such effects and the underlying molecular mechanisms remain incompletely understood. Methods PBMC and monocytes were isolated from healthy donors and in vitro stimulated with V132, MV140 or MV140/V132. After 6 days of resting, cells were reestimulated with LPS and MV140. Analysis of cytokine production by ELISA, Seahorse assays for functional metabolic experiments and chromatin immunoprecipitation assays were performed. BALB/c mice were intraperitoneally and sublingually immunized with V132. Results We uncover that V132 induces trained immunity in human PBMCs and purified monocytes, significantly increasing the responses triggered by subsequent stimulation with MV140. Mechanistically, V132 drives metabolic rewiring towards increased glycolysis and oxidative phosphorylation and induces epigenetic reprogramming that enhances the transcription of the pro-inflammatory genes IL6 and TNFA. Splenocytes and peritoneal cells from V132-immunize mice show increased responses upon in vitro stimulation with MV140. Remarkably, splenocytes from sublingually V132-immunized and MV140 in vivo treatment mice show stronger Th17 responses than mice exposed to excipients upon in vitro stimulation with MV140. Conclusion Overall, we provide novel mechanistic insights into how V132-induced trained immunity enhances both innate and adaptive immune responses triggered by MV140, which might open the door for new interventions for GUTIs with important clinical implications.
Collapse
Affiliation(s)
- Leticia Martín-Cruz
- Department of Biochemistry and Molecular Biology, School of Chemistry, Complutense University of Madrid, Madrid, Spain
| | - Alba Angelina
- Department of Biochemistry and Molecular Biology, School of Chemistry, Complutense University of Madrid, Madrid, Spain
| | - Ilayda Baydemir
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Özlem Bulut
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Centre, Nijmegen, Netherlands
| | | | - Mihai G. Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Centre, Nijmegen, Netherlands
- Department of Immunology and Metabolism, Life and Medical Sciences Institute, University of Bonn, Bonn, Germany
| | - Jorge Domínguez-Andrés
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Oscar Palomares
- Department of Biochemistry and Molecular Biology, School of Chemistry, Complutense University of Madrid, Madrid, Spain
| |
Collapse
|
476
|
Sunoqrot S, Al-Bakri AG, Ibrahim LH, Aldaken N. Amphotericin B-Loaded Plant-Inspired Polyphenol Nanoparticles Enhance Its Antifungal Activity and Biocompatibility. ACS APPLIED BIO MATERIALS 2022; 5:5156-5164. [PMID: 36241585 DOI: 10.1021/acsabm.2c00537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Amphotericin B (AmB) is one of the first-line treatments for systemic fungal infections, yet it suffers from dose-limiting systemic toxicity and high cost of less toxic lipid-based formulations. Here, we report on a facile approach to synthesize an AmB-loaded nanomedicine by leveraging plant-inspired oxidative self-polymerization of the ubiquitous polyphenol quercetin (QCT). Polymerized QCT nanoparticles (pQCT NPs) were formed, loaded with AmB, and functionalized with poly(ethylene glycol) (PEG) to impart steric stability in a simple procedure that relied on mixing followed by dialysis. The AmB-loaded NPs (AmB@pQCT-PEG NPs) were characterized by a drug loading efficiency of more than 90%, a particle size of around 160 nm, a polydispersity index of 0.07, and a partially negative surface charge. AmB release from the NPs was sustained over several days and followed the Korsmeyer-Peppas model with a release exponent (n) value >0.85, denoting drug release by polymer relaxation and swelling. A hemolysis assay revealed the NPs to be highly biocompatible, with negligible hemolytic activity and 30-60% hemolysis after 1 and 24 h of incubation with erythrocytes, respectively, across a wide concentration range (6.25-100.00 μg/mL). Conversely, equivalent concentrations of free AmB caused 90-100% hemolysis within the same timeframe. Importantly, AmB@pQCT-PEG NPs outperformed free AmB in microbial susceptibility assays on Candida albicans, achieving a minimum inhibitory concentration of 62.5 ng/mL after 48 h of incubation, which was 2-fold lower than the free drug. Our results demonstrate that pQCT NPs may serve as a viable AmB delivery platform for the treatment of fungal infections and potentially other AmB-susceptible pathogens.
Collapse
Affiliation(s)
- Suhair Sunoqrot
- Department of Pharmacy, Faculty of Pharmacy, Al-Zaytoonah University of Jordan, Amman11733, Jordan
| | - Amal G Al-Bakri
- Department of Pharmaceutics and Pharmaceutical Technology, School of Pharmacy, The University of Jordan, Amman11942, Jordan
| | - Lina Hasan Ibrahim
- Department of Pharmacy, Faculty of Pharmacy, Al-Zaytoonah University of Jordan, Amman11733, Jordan
| | - Neda'a Aldaken
- Department of Pharmaceutics and Pharmaceutical Technology, School of Pharmacy, The University of Jordan, Amman11942, Jordan
| |
Collapse
|
477
|
Teixeira MM, Carvalho DT, Sousa E, Pinto E. New Antifungal Agents with Azole Moieties. Pharmaceuticals (Basel) 2022; 15:1427. [PMID: 36422557 PMCID: PMC9698508 DOI: 10.3390/ph15111427] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 11/08/2022] [Accepted: 11/14/2022] [Indexed: 09/22/2023] Open
Abstract
Fungal conditions affect a multitude of people worldwide, leading to increased hospitalization and mortality rates, and the need for novel antifungals is emerging with the rise of resistance and immunocompromised patients. Continuous use of azole drugs, which act by inhibiting the fungal CYP51, involved in the synthesis of ergosterol, essential to the fungal cell membrane, has enhanced the resistance and tolerance of some fungal strains to treatment, thereby limiting the arsenal of available drugs. The goal of this review is to gather literature information on new promising azole developments in clinical trials, with in vitro and in vivo results against fungal strains, and complementary assays, such as toxicity, susceptibility assays, docking studies, among others. Several molecules are reviewed as novel azole structures in clinical trials and with recent/imminent approvals, as well as other innovative molecules with promising antifungal activity. Structure-activity relationship (SAR) studies are displayed whenever possible. The azole moiety is brought over as a privileged structure, with multiple different compounds emerging with distinct pharmacophores and SAR. Particularly, 1,2,3-triazole natural product conjugates emerged in the last years, presenting promising antifungal activity and a broad spectrum against various fungi.
Collapse
Affiliation(s)
- Melissa Martins Teixeira
- Laboratory of Organic and Pharmaceutical Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
- Laboratory of Microbiology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR), University of Porto, 4450-208 Matosinhos, Portugal
| | - Diogo Teixeira Carvalho
- Laboratory of Organic and Pharmaceutical Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
- Laboratory of Research in Pharmaceutical Chemistry, Department of Food and Drugs, Faculty of Pharmaceutical Sciences, Federal University of Alfenas, Alfenas 37137-001, Brazil
| | - Emília Sousa
- Laboratory of Organic and Pharmaceutical Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR), University of Porto, 4450-208 Matosinhos, Portugal
| | - Eugénia Pinto
- Laboratory of Microbiology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR), University of Porto, 4450-208 Matosinhos, Portugal
| |
Collapse
|
478
|
Nelson BN, Daugherty CS, Sharp RR, Booth JL, Patel VI, Metcalf JP, Jones KL, Wozniak KL. Protective interaction of human phagocytic APC subsets with Cryptococcus neoformans induces genes associated with metabolism and antigen presentation. Front Immunol 2022; 13:1054477. [PMID: 36466930 PMCID: PMC9709479 DOI: 10.3389/fimmu.2022.1054477] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 10/25/2022] [Indexed: 09/01/2023] Open
Abstract
Cryptococcal meningitis is the most common cause of meningitis among HIV/AIDS patients in sub-Saharan Africa, and worldwide causes over 223,000 cases leading to more than 181,000 annual deaths. Usually, the fungus gets inhaled into the lungs where the initial interactions occur with pulmonary phagocytes such as dendritic cells and macrophages. Following phagocytosis, the pathogen can be killed or can replicate intracellularly. Previous studies in mice showed that different subsets of these innate immune cells can either be antifungal or permissive for intracellular fungal growth. Our studies tested phagocytic antigen-presenting cell (APC) subsets from the human lung against C. neoformans. Human bronchoalveolar lavage was processed for phagocytic APCs and incubated with C. neoformans for two hours to analyze the initial interactions and fate of the fungus, living or killed. Results showed all subsets (3 macrophage and 3 dendritic cell subsets) interacted with the fungus, and both living and killed morphologies were discernable within the subsets using imaging flow cytometry. Single cell RNA-seq identified several different clusters of cells which more closely related to interactions with C. neoformans and its protective capacity against the pathogen rather than discrete cellular subsets. Differential gene expression analyses identified several changes in the innate immune cell's transcriptome as it kills the fungus including increases of TNF-α (TNF) and the switch to using fatty acid metabolism by upregulation of the gene FABP4. Also, increases of TNF-α correlated to cryptococcal interactions and uptake. Together, these analyses implicated signaling networks that regulate expression of many different genes - both metabolic and immune - as certain clusters of cells mount a protective response and kill the pathogen. Future studies will examine these genes and networks to understand the exact mechanism(s) these phagocytic APC subsets use to kill C. neoformans in order to develop immunotherapeutic strategies to combat this deadly disease.
Collapse
Affiliation(s)
- Benjamin N. Nelson
- Department of Microbiology and Molecular Genetics, Oklahoma State University, Stillwater, OK, United States
| | - Cheyenne S. Daugherty
- Department of Microbiology and Molecular Genetics, Oklahoma State University, Stillwater, OK, United States
| | - Rachel R. Sharp
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - J. Leland Booth
- Department of Medicine, Pulmonary, Critical Care & Sleep Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Vineet I. Patel
- Department of Medicine, Pulmonary, Critical Care & Sleep Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Jordan P. Metcalf
- Department of Medicine, Pulmonary, Critical Care & Sleep Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- Veterans Affairs Medical Center, Oklahoma City, OK, United States
| | - Kenneth L. Jones
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Karen L. Wozniak
- Department of Microbiology and Molecular Genetics, Oklahoma State University, Stillwater, OK, United States
| |
Collapse
|
479
|
Townsend L, Martin-Loeches I. Invasive Aspergillosis in the Intensive Care Unit. Diagnostics (Basel) 2022; 12:2712. [PMID: 36359555 PMCID: PMC9689891 DOI: 10.3390/diagnostics12112712] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 11/02/2022] [Accepted: 11/04/2022] [Indexed: 08/28/2023] Open
Abstract
Invasive pulmonary aspergillosis (IPA) is a serious condition resulting in significant mortality and morbidity among patients in intensive care units (ICUs). There is a growing number of at-risk patients for this condition with the increasing use of immunosuppressive therapies. The diagnosis of IPA can be difficult in ICUs, and relies on integration of clinical, radiological, and microbiological features. In this review, we discuss patient populations at risk for IPA, as well as the diagnostic criteria employed. We review the fungal biomarkers used, as well as the challenges in distinguishing colonization with Aspergillus from invasive disease. We also address the growing concern of multidrug-resistant Aspergillosis and review the new and novel therapeutics which are in development to combat this.
Collapse
Affiliation(s)
- Liam Townsend
- Department of Intensive Care Medicine, Multidisciplinary Intensive Care Research Organization (MICRO), St. James’s Hospital, D08 NHY1 Dublin, Ireland
| | - Ignacio Martin-Loeches
- Department of Intensive Care Medicine, Multidisciplinary Intensive Care Research Organization (MICRO), St. James’s Hospital, D08 NHY1 Dublin, Ireland
- Department of Clinical Medicine, School of Medicine, Trinity College Dublin, D02 PN91 Dublin, Ireland
- Hospital Clinic, Institut D’Investigacions Biomediques August Pi i Sunyer (IDIBAPS), Universidad de Barcelona, Ciberes, 08036 Barcelona, Spain
| |
Collapse
|
480
|
Antimicrobial stewardship in the post COVID-19 pandemic era: an opportunity for renewed focus on controlling the threat of antimicrobial resistance. J Hosp Infect 2022; 129:121-123. [PMID: 36280374 PMCID: PMC9585509 DOI: 10.1016/j.jhin.2022.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
481
|
Frei A, Elliott AG, Kan A, Dinh H, Bräse S, Bruce AE, Bruce MR, Chen F, Humaidy D, Jung N, King AP, Lye PG, Maliszewska HK, Mansour AM, Matiadis D, Muñoz MP, Pai TY, Pokhrel S, Sadler PJ, Sagnou M, Taylor M, Wilson JJ, Woods D, Zuegg J, Meyer W, Cain AK, Cooper MA, Blaskovich MAT. Metal Complexes as Antifungals? From a Crowd-Sourced Compound Library to the First In Vivo Experiments. JACS AU 2022; 2:2277-2294. [PMID: 36311838 PMCID: PMC9597602 DOI: 10.1021/jacsau.2c00308] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 07/01/2022] [Accepted: 07/27/2022] [Indexed: 06/16/2023]
Abstract
There are currently fewer than 10 antifungal drugs in clinical development, but new fungal strains that are resistant to most current antifungals are spreading rapidly across the world. To prevent a second resistance crisis, new classes of antifungal drugs are urgently needed. Metal complexes have proven to be promising candidates for novel antibiotics, but so far, few compounds have been explored for their potential application as antifungal agents. In this work, we report the evaluation of 1039 metal-containing compounds that were screened by the Community for Open Antimicrobial Drug Discovery (CO-ADD). We show that 20.9% of all metal compounds tested have antimicrobial activity against two representative Candida and Cryptococcus strains compared with only 1.1% of the >300,000 purely organic molecules tested through CO-ADD. We identified 90 metal compounds (8.7%) that show antifungal activity while not displaying any cytotoxicity against mammalian cell lines or hemolytic properties at similar concentrations. The structures of 21 metal complexes that display high antifungal activity (MIC ≤1.25 μM) are discussed and evaluated further against a broad panel of yeasts. Most of these have not been previously tested for antifungal activity. Eleven of these metal complexes were tested for toxicity in the Galleria mellonella moth larva model, revealing that only one compound showed signs of toxicity at the highest injected concentration. Lastly, we demonstrated that the organo-Pt(II) cyclooctadiene complex Pt1 significantly reduces fungal load in an in vivo G. mellonella infection model. These findings showcase that the structural and chemical diversity of metal-based compounds can be an invaluable tool in the development of new drugs against infectious diseases.
Collapse
Affiliation(s)
- Angelo Frei
- Centre
for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland4072, Australia
- Department
of Chemistry, Biochemistry & Pharmaceutical Sciences, University of Bern, Freiestrasse 3, 3012Bern, Switzerland
| | - Alysha G. Elliott
- Centre
for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland4072, Australia
| | - Alex Kan
- Molecular
Mycology Research Laboratory, Centre for Infectious Diseases and Microbiology,
Faculty of Medicine and Health, Sydney Medical School, Westmead Clinical
School, Sydney Institute for Infectious Diseases, Westmead Hospital-Research
and Education Network, Westmead Institute for Medical Research, University of Sydney, Sydney, NSW2145, Australia
| | - Hue Dinh
- School
of Natural Sciences, ARC Centre of Excellence in Synthetic Biology, Macquarie University, Sydney, NSW2109, Australia
| | - Stefan Bräse
- Institute
of Organic Chemistry, Karlsruhe Institute
of Technology, Fritz-Haber-Weg 6, 76131Karlsruhe, Germany
- Institute
of Biological and Chemical Systems - Functional Molecular Systems, Karlsruhe Institute of Technology, 76344Eggenstein-Leopoldshafen, Germany
| | - Alice E. Bruce
- Department
of Chemistry, University of Maine, Orono, Maine04469, United States
| | - Mitchell R. Bruce
- Department
of Chemistry, University of Maine, Orono, Maine04469, United States
| | - Feng Chen
- Department
of Chemistry, University of Warwick, Gibbet Hill Road, CoventryCV4 7AL, U.K.
| | - Dhirgam Humaidy
- Department
of Chemistry, University of Maine, Orono, Maine04469, United States
| | - Nicole Jung
- Karlsruhe
Nano Micro Facility (KNMF), Karlsruhe Institute
of Technology, Hermann-von-Helmholtz-Platz 1, 76344Eggenstein-Leopoldshafen, Germany
- Institute
of Biological and Chemical Systems - Functional Molecular Systems, Karlsruhe Institute of Technology, 76344Eggenstein-Leopoldshafen, Germany
| | - A. Paden King
- Department
of Chemistry and Chemical Biology, Cornell
University, Ithaca, New York14853, United States
| | - Peter G. Lye
- School
of Science and Technology, University of
New England, Armidale, NSW2351, Australia
| | - Hanna K. Maliszewska
- School
of Chemistry, University of East Anglia, Norwich Research Park, NorwichNR4 7TJ, U.K.
| | - Ahmed M. Mansour
- Chemistry
Department, Faculty of Science, Cairo University, Giza12613, Egypt
| | - Dimitris Matiadis
- Institute
of Biosciences & Applications, National
Centre for Scientific Research “Demokritos”, 15310Athens, Greece
| | - María Paz Muñoz
- School
of Chemistry, University of East Anglia, Norwich Research Park, NorwichNR4 7TJ, U.K.
| | - Tsung-Yu Pai
- Molecular
Mycology Research Laboratory, Centre for Infectious Diseases and Microbiology,
Faculty of Medicine and Health, Sydney Medical School, Westmead Clinical
School, Sydney Institute for Infectious Diseases, Westmead Hospital-Research
and Education Network, Westmead Institute for Medical Research, University of Sydney, Sydney, NSW2145, Australia
| | - Shyam Pokhrel
- Department
of Chemistry, University of Maine, Orono, Maine04469, United States
| | - Peter J. Sadler
- Department
of Chemistry, University of Warwick, Gibbet Hill Road, CoventryCV4 7AL, U.K.
| | - Marina Sagnou
- Institute
of Biosciences & Applications, National
Centre for Scientific Research “Demokritos”, 15310Athens, Greece
| | - Michelle Taylor
- School
of Science and Technology, University of
New England, Armidale, NSW2351, Australia
| | - Justin J. Wilson
- Department
of Chemistry and Chemical Biology, Cornell
University, Ithaca, New York14853, United States
| | - Dean Woods
- School
of Science and Technology, University of
New England, Armidale, NSW2351, Australia
| | - Johannes Zuegg
- Centre
for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland4072, Australia
| | - Wieland Meyer
- Molecular
Mycology Research Laboratory, Centre for Infectious Diseases and Microbiology,
Faculty of Medicine and Health, Sydney Medical School, Westmead Clinical
School, Sydney Institute for Infectious Diseases, Westmead Hospital-Research
and Education Network, Westmead Institute for Medical Research, University of Sydney, Sydney, NSW2145, Australia
| | - Amy K. Cain
- School
of Natural Sciences, ARC Centre of Excellence in Synthetic Biology, Macquarie University, Sydney, NSW2109, Australia
| | - Matthew A. Cooper
- Centre
for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland4072, Australia
| | - Mark A. T. Blaskovich
- Centre
for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland4072, Australia
| |
Collapse
|
482
|
Sprute R, Grothe JH, Heringer S, Cornely OA. Reason and reality-identifying barriers to patient enrolment for clinical trials in invasive candidiasis. J Antimicrob Chemother 2022; 77:3475-3481. [PMID: 36214051 DOI: 10.1093/jac/dkac344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Accepted: 09/19/2022] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVES Enrolment of subjects to clinical trials investigating novel drugs for infectious diseases is an ongoing challenge. In this study, we evaluate factors associated with non-enrolment in treatment trials for invasive candidiasis. METHODS We conducted a retrospective review of pre-screening logs of patients that were assessed for enrolment in the three clinical trials ACTIVE (NCT00413218), APX001-201 (NCT03604705) and ReSTORE (NCT03667690), investigating novel drugs for invasive candidiasis between September 2007 and August 2021 to identify reasons for study ineligibility. RESULTS Two hundred and fifty-six patients with invasive candidiasis were identified for potential study participation with n = 154 for the ACTIVE trial, n = 89 for APX001-201 and n = 13 for ReSTORE. Half of the potential participants were unable or unwilling to consent. We further identified comorbid conditions such as hepatic or renal impairment [21 hepatic and renal cases (13.6%) in ACTIVE; 12 hepatic (13.5%) and 28 renal cases (31.5%) in APX], prior antifungal treatment [11 cases (7.1%) in ACTIVE; 16 (18.0%) in APX; 7 (38.5%) in ReSTORE] and the last positive culture obtained ≥96 h prior to dosing [1 case (0.6%) in ACTIVE; 7 (7.9%) in APX; 5 (38.5%) in ReSTORE] as relevant reasons for non-enrolment. We also identified criteria repetitively used in the analysed studies that did not contribute substantially to ineligibility rates. Ultimately, 254/256 patients (99.2%) were ineligible for enrolment in the respective trial. CONCLUSIONS This study identified barriers to enrolment in clinical trials assessing novel antifungal agents in invasive candidiasis. Identification of eligibility criteria associated with non-enrolment allows modification of future trial designs and may ultimately result in higher recruitment rates.
Collapse
Affiliation(s)
- Rosanne Sprute
- Department I of Internal Medicine, University of Cologne, Faculty of Medicine and University Hospital Cologne, Excellence Center for Medical Mycology (ECMM), Cologne, NRW, Germany.,University of Cologne, Faculty of Medicine and University Hospital Cologne, Chair Translational Research, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne, NRW, Germany.,German Centre for Infection Research (DZIF), Partner Site Bonn-Cologne, Cologne, NRW, Germany
| | - Jan H Grothe
- Department I of Internal Medicine, University of Cologne, Faculty of Medicine and University Hospital Cologne, Excellence Center for Medical Mycology (ECMM), Cologne, NRW, Germany.,University of Cologne, Faculty of Medicine and University Hospital Cologne, Chair Translational Research, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne, NRW, Germany.,German Centre for Infection Research (DZIF), Partner Site Bonn-Cologne, Cologne, NRW, Germany
| | - Sarah Heringer
- Department I of Internal Medicine, University of Cologne, Faculty of Medicine and University Hospital Cologne, Excellence Center for Medical Mycology (ECMM), Cologne, NRW, Germany.,University of Cologne, Faculty of Medicine and University Hospital Cologne, Chair Translational Research, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne, NRW, Germany.,German Centre for Infection Research (DZIF), Partner Site Bonn-Cologne, Cologne, NRW, Germany
| | - Oliver A Cornely
- Department I of Internal Medicine, University of Cologne, Faculty of Medicine and University Hospital Cologne, Excellence Center for Medical Mycology (ECMM), Cologne, NRW, Germany.,University of Cologne, Faculty of Medicine and University Hospital Cologne, Chair Translational Research, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne, NRW, Germany.,German Centre for Infection Research (DZIF), Partner Site Bonn-Cologne, Cologne, NRW, Germany.,University of Cologne, Faculty of Medicine and University Hospital Cologne, Clinical Trials Centre Cologne (ZKS Köln), Cologne, NRW, Germany.,University of Cologne, Faculty of Medicine and University Hospital Cologne, Center for Molecular Medicine Cologne (CMMC), Cologne, Germany
| |
Collapse
|
483
|
Huang M, Yang L, Zhou L, Sun C, Zhao W, Peng J, Jiao Z, Tian C, Guo G. Identification and functional characterization of ORF19.5274, a novel gene involved in both azoles susceptibility and hypha development in Candida albicans. Front Microbiol 2022; 13:990318. [PMID: 36262330 PMCID: PMC9575988 DOI: 10.3389/fmicb.2022.990318] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Accepted: 08/26/2022] [Indexed: 11/13/2022] Open
Abstract
Azole resistance is becoming increasingly serious due to the frequent recurrence of fungal infections and the need for long-term clinical prevention. In our previous study, we discovered ORF19.5274 with an unknown function by TMT™ quantitative proteomics technology after fluconazole (FLC) treatment of Candida albicans. In this study, we created the target gene deletion strain using CRISPR-Cas9 editing technology to see if ORF19.5274 regulates azole sensitivity. The data showed that ORF19.5274 was involved in hyphal development and susceptibility to antifungal azoles. Deleting this gene resulted in defective hyphal growth in solid medium, while only a weak lag in the initiation of hyphal development and restoring hyphal growth during the hyphal maintenance phase under liquid conditions. Moreover, intracellular reactive oxygen species (ROS) assay and propidium iodide staining assays showed increased endogenous ROS levels and membrane permeability, but decreased metabolic activity of biofilm in orf19.5274Δ/Δ after treatment with FLC in comparison with either SC5314 or orf19.5274Δ/Δ::ORF19.5274 strains. More importantly, orf19.5274Δ/Δ significantly enhanced the FLC efficacy against C. albicans in infected Galleria mellonella larvae. The above characteristics were fully or partially restored in the complemented strain indicating that the changes caused by ORF19.5274 deletion were specific. In summary, the ORF19.5274 gene is required for hyphal development of C. albicans, and is correlated with the response to antifungal azoles in vitro and in vivo. The identification of ORF19.5275 is promising to expand the potential candidate targets for azoles.
Collapse
Affiliation(s)
- Mingjiao Huang
- The Key and Characteristic Laboratory of Modern Pathogen Biology, School of Basic Medical Sciences, Guizhou Medical University, Guiyang, China
| | - Longbing Yang
- The Key and Characteristic Laboratory of Modern Pathogen Biology, School of Basic Medical Sciences, Guizhou Medical University, Guiyang, China
| | - Luoxiong Zhou
- The Key and Characteristic Laboratory of Modern Pathogen Biology, School of Basic Medical Sciences, Guizhou Medical University, Guiyang, China
- Key Laboratory of Environmental Pollution Monitoring and Disease Control (Guizhou Medical University), Ministry of Education, Guiyang, China
| | - Chaoqin Sun
- The Key and Characteristic Laboratory of Modern Pathogen Biology, School of Basic Medical Sciences, Guizhou Medical University, Guiyang, China
- Center of Laboratory Medicine, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Wenjing Zhao
- The Key and Characteristic Laboratory of Modern Pathogen Biology, School of Basic Medical Sciences, Guizhou Medical University, Guiyang, China
| | - Jian Peng
- The Key and Characteristic Laboratory of Modern Pathogen Biology, School of Basic Medical Sciences, Guizhou Medical University, Guiyang, China
- Key Laboratory of Environmental Pollution Monitoring and Disease Control (Guizhou Medical University), Ministry of Education, Guiyang, China
| | - Zhenlong Jiao
- The Key and Characteristic Laboratory of Modern Pathogen Biology, School of Basic Medical Sciences, Guizhou Medical University, Guiyang, China
- Translational Medicine Research Center, Guizhou Medical University, Guiyang, China
| | - Chunren Tian
- The Key and Characteristic Laboratory of Modern Pathogen Biology, School of Basic Medical Sciences, Guizhou Medical University, Guiyang, China
| | - Guo Guo
- The Key and Characteristic Laboratory of Modern Pathogen Biology, School of Basic Medical Sciences, Guizhou Medical University, Guiyang, China
- Key Laboratory of Environmental Pollution Monitoring and Disease Control (Guizhou Medical University), Ministry of Education, Guiyang, China
- Translational Medicine Research Center, Guizhou Medical University, Guiyang, China
- *Correspondence: Guo Guo,
| |
Collapse
|
484
|
Langfeldt A, Gold JAW, Chiller T. Emerging Fungal Infections: from the Fields to the Clinic, Resistant Aspergillus fumigatus and Dermatophyte Species: a One Health Perspective on an Urgent Public Health Problem. CURRENT CLINICAL MICROBIOLOGY REPORTS 2022; 9:46-51. [PMID: 36188157 PMCID: PMC9512973 DOI: 10.1007/s40588-022-00181-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/16/2022] [Indexed: 11/27/2022]
Abstract
Purpose of Review For this review, we use a One Health approach to examine two globally emerging public health threats related to antifungal drug resistance: triazole-resistant Aspergillus fumigatus infections, which can cause a life-threatening illness in immunocompromised hosts, and antifungal-resistant dermatophytosis, which is an aggressive skin infection caused by dermatophyte molds. We describe the state of current scientific knowledge and outline necessary public health actions to address each issue. Recent Findings Recent evidence has identified the agricultural use of triazole fungicides as an important driver of triazole-resistant A. fumigatus infections. Antifungal-resistant dermatophyte infections are likely driven by the inappropriate use of antifungal drugs and antibacterial and corticosteroid creams. Summary This review highlights the need for a One Health approach to address emerging antifungal resistant infections, emphasizing judicious antifungal use to preserve available treatments; strengthened laboratory capacity to identify antifungal resistance; and improved human, animal, and environmental surveillance to detect emerging resistance, monitor trends, and evaluate the effectiveness of efforts to decrease spread.
Collapse
Affiliation(s)
- Antonia Langfeldt
- Mycotic Diseases Branch, Division of Foodborne, Waterborne, and Environmental Diseases, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, GA USA
| | - Jeremy A. W. Gold
- Mycotic Diseases Branch, Division of Foodborne, Waterborne, and Environmental Diseases, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, GA USA
| | - Tom Chiller
- Mycotic Diseases Branch, Division of Foodborne, Waterborne, and Environmental Diseases, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, GA USA
| |
Collapse
|
485
|
Canh Pham E, Truong TN. Design, Microwave-Assisted Synthesis, Antimicrobial and Anticancer Evaluation, and In Silico Studies of Some 2-Naphthamide Derivatives as DHFR and VEGFR-2 Inhibitors. ACS OMEGA 2022; 7:33614-33628. [PMID: 36157776 PMCID: PMC9494668 DOI: 10.1021/acsomega.2c05206] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Accepted: 08/31/2022] [Indexed: 06/16/2023]
Abstract
Naphthamide is a common structural framework with diverse pharmacological activities. Ten novel 2-naphthamide derivatives have been designed, synthesized, and evaluated for their in vitro antibacterial, antifungal, and anticancer activities. The title compounds were synthesized from dimethoxybenzaldehyde derivatives through a four-step microwave-assisted synthesis process. The structures were confirmed by 1H NMR, 13C NMR, and MS spectra. Compound 8b showed good antibacterial activity against Escherichia coli, Streptococcus faecalis, Salmonella enterica, MSSA, and MRSA with MIC values of 16, 16, 16, 8, and 16 μg/mL, respectively, compared to ciprofloxacin (MIC = 8-16 μg/mL). Compounds 5b (IC50 = 3.59-8.38 μM) and 8b (IC50 = 2.97-7.12 μM) exhibited good cytotoxic activity against C26, HepG2, and MCF7 cancer cell lines as compared to paclitaxel (IC50 = 2.85-5.75 μM). Moreover, compounds 5b and 8b exhibited better anticancer activity than PTX against the C26 cell line. In particular, compound 8b showed potent in vitro VEGFR-2 inhibitory activity with the IC50 value of 0.384 μM compared with sorafenib (IC50 = 0.069 μM). Therefore, compound 8b is the most potent compound for anticancer activity as indicated by in vitro cell line inhibition, in silico ADMET, molecular docking, and in vitro VEGFR-2 inhibition studies.
Collapse
Affiliation(s)
- Em Canh Pham
- Department
of Medicinal Chemistry, Faculty of Pharmacy, Hong Bang International University, 700000 Ho Chi Minh City, Vietnam
| | - Tuyen Ngoc Truong
- Department
of Organic Chemistry, Faculty of Pharmacy, University of Medicine and Pharmacy at Ho Chi Minh City, 700000 Ho Chi Minh
City, Vietnam
| |
Collapse
|
486
|
Penicillium digitatum as a Model Fungus for Detecting Antifungal Activity of Botanicals: An Evaluation on Vietnamese Medicinal Plant Extracts. J Fungi (Basel) 2022; 8:jof8090956. [PMID: 36135681 PMCID: PMC9502062 DOI: 10.3390/jof8090956] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 09/05/2022] [Accepted: 09/11/2022] [Indexed: 11/17/2022] Open
Abstract
Medicinal plants play important roles in traditional medicine, and numerous compounds among them have been recognized for their antimicrobial activity. However, little is known about the potential of Vietnamese medicinal plants for antifungal activity. In this study, we examined the antagonistic activity of twelve medicinal plant species collected in Northern Vietnam against Penicillium digitatum, Aspergillus flavus, Aspergillus fumigatus, and Candida albicans. The results showed that the antifungal activities of the crude extracts from Mahonia bealei, Ficus semicordata, and Gnetum montanum were clearly detected with the citrus postharvest pathogen P. digitatum. These extracts could fully inhibit the growth of P. digitatum on the agar medium, and on the infected citrus fruits at concentrations of 300–1000 µg/mL. Meanwhile, the other tested fungi were less sensitive to the antagonistic activity of the plant extracts. In particular, we found that the ethanolic extract of M. bealei displayed a broad-spectrum antifungal activity against all four pathogenic fungi. Analysis of this crude extract by enrichment coupled with high-performance liquid chromatography revealed that berberine and palmatine are major metabolites. Additional inspections indicated berberine as the key compound responsible for the antifungal activity of the M. bealei ethanolic extract. Our study provides a better understanding of the potential of Vietnamese medicinal plant resources for combating fungal pathogens. This work also highlights that the citrus pathogen P. digitatum can be employed as a model fungus for screening the antifungal activity of botanicals.
Collapse
|
487
|
Gow NAR, Johnson C, Berman J, Coste AT, Cuomo CA, Perlin DS, Bicanic T, Harrison TS, Wiederhold N, Bromley M, Chiller T, Edgar K. The importance of antimicrobial resistance in medical mycology. Nat Commun 2022; 13:5352. [PMID: 36097014 PMCID: PMC9466305 DOI: 10.1038/s41467-022-32249-5] [Citation(s) in RCA: 100] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 07/22/2022] [Indexed: 01/08/2023] Open
Abstract
Prior to the SARS-CoV-2 pandemic, antibiotic resistance was listed as the major global health care priority. Some analyses, including the O'Neill report, have predicted that deaths due to drug-resistant bacterial infections may eclipse the total number of cancer deaths by 2050. Although fungal infections remain in the shadow of public awareness, total attributable annual deaths are similar to, or exceeds, global mortalities due to malaria, tuberculosis or HIV. The impact of fungal infections has been exacerbated by the steady rise of antifungal drug resistant strains and species which reflects the widespread use of antifungals for prophylaxis and therapy, and in the case of azole resistance in Aspergillus, has been linked to the widespread agricultural use of antifungals. This review, based on a workshop hosted by the Medical Research Council and the University of Exeter, illuminates the problem of antifungal resistance and suggests how this growing threat might be mitigated.
Collapse
Affiliation(s)
- Neil A R Gow
- MRC Centre for Medical Mycology, School of Biosciences, University of Exeter, Geoffrey Pope Building, Exeter, EX4 4QD, UK.
| | - Carolyn Johnson
- Medical Research Council, Polaris House, Swindon, SN2 1FL, UK.
| | - Judith Berman
- Shmunis School of Biomedical and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, 418 Britannia Building, Ramat Aviv, 69978, Israel
| | - Alix T Coste
- Microbiology Institute, University Hospital Lausanne, rue du Bugnon 48, 1011, Lausanne, Switzerland
| | - Christina A Cuomo
- (CAC) Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - David S Perlin
- Center for Discovery and Innovation, Hackensack Meridian health, Nutley, NJ, 07110, USA
| | - Tihana Bicanic
- Institute of Infection and Immunity, St George's University of London, London, SW17 0RE, UK
- Clinical Academic Group in Infection, St George's University Hospitals NHS Foundation Trust, London, SW17 0QT, UK
| | - Thomas S Harrison
- MRC Centre for Medical Mycology, School of Biosciences, University of Exeter, Geoffrey Pope Building, Exeter, EX4 4QD, UK
- Institute of Infection and Immunity, St George's University of London, London, SW17 0RE, UK
- Clinical Academic Group in Infection, St George's University Hospitals NHS Foundation Trust, London, SW17 0QT, UK
| | - Nathan Wiederhold
- Department of Pathology and Laboratory Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Mike Bromley
- Manchester Fungal Infection Group, Division of Evolution, Infection, and Genomics, Faculty of Biology, Medicine and Health, University of Manchester, CTF Building, 46 Grafton Street, Manchester, M13 9NT, UK
| | - Tom Chiller
- Center for Disease Control and Prevention Mycotic Disease Branch 1600 Clifton Rd, MSC-09, Atlanta, 30333, GA, USA
| | - Keegan Edgar
- Center for Disease Control and Prevention Mycotic Disease Branch 1600 Clifton Rd, MSC-09, Atlanta, 30333, GA, USA
| |
Collapse
|
488
|
Xu J. Assessing global fungal threats to humans. MLIFE 2022; 1:223-240. [PMID: 38818220 PMCID: PMC10989982 DOI: 10.1002/mlf2.12036] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Revised: 08/02/2022] [Accepted: 08/06/2022] [Indexed: 06/01/2024]
Abstract
Fungi are an integral part of the earth's biosphere. They are broadly distributed in all continents and ecosystems and play a diversity of roles. Here, I review our current understanding of fungal threats to humans and describe the major factors that contribute to various threats. Among the 140,000 or so known species out of the estimated six million fungal species on Earth, about 10% directly or indirectly threaten human health and welfare. Major threats include mushroom poisoning, fungal allergies, infections of crop plants, food contamination by mycotoxins, and mycoses in humans. A growing number of factors have been identified to impact various fungal threats, including human demographics, crop distributions, anthropogenic activities, pathogen dispersals, global climate change, and/or the applications of antifungal drugs and agricultural fungicides. However, while models have been developed for analyzing various processes of individual threats and threat managements, current data are primarily descriptive and incomplete, and there are significant obstacles to integration of the diverse factors into accurate quantitative assessments of fungal threats. With increasing technological advances and concerted efforts to track the spatial and temporal data on climate and environmental variables; mycotoxins in the feed and food supply chains; fungal population dynamics in crop fields, human and animal populations, and the environment; human population demographics; and the prevalence and severities of fungal allergies and diseases, our ability to accurately assess fungal threats will improve. Such improvements should help us develop holistic strategies to manage fungal threats in the future.
Collapse
Affiliation(s)
- Jianping Xu
- Department of Biology and Institute of Infectious Diseases ResearchMcMaster UniversityHamiltonOntarioCanada
| |
Collapse
|
489
|
The Antifungal and Antibiofilm Activities of Caffeine against Candida albicans on Polymethyl Methacrylate Denture Base Material. Biomedicines 2022; 10:biomedicines10092078. [PMID: 36140179 PMCID: PMC9495344 DOI: 10.3390/biomedicines10092078] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 08/17/2022] [Accepted: 08/23/2022] [Indexed: 11/18/2022] Open
Abstract
Background: In this study, the effect of pure caffeine was established against Candida albicans (C. albicans) using different microbiological techniques. Methods: Broth microdilution and colony forming units (CFUs) assays were used to detect the minimum inhibitory concentration (MIC) and minimum fungicidal concentration (MFC). The Live/Dead fluorescent dyes were implemented to determine the yeast viability. Polymethyl methacrylate acrylic resin (PMMA) discs were prepared to evaluate caffeine’s effects against adherent C. albicans using microplate reader, CFUs, and scanning electron microscope (SEM). Results: caffeine’s MIC was detected around 30 mg/mL, while the MFC was considered at 60 mg/mL. In an agar-well diffusion test, the inhibition zones were wider in caffeine groups. The Live/Dead viability test verified caffeine’s antifungal effects. The optical density of the adherent C. albicans on PMMA discs were lower at 620 nm or 410 nm in caffeine groups. CFU count was also reduced by caffeine treatments. SEM revealed the lower adherent C. albicans count in caffeine groups. The effect of caffeine was dose-dependent at which the 60 mg/mL dose demonstrated the most prominent effect. Conclusion: The study reinforced caffeine’s antifungal and antibiofilm properties and suggested it as an additive, or even an alternative, disinfectant solution for fungal biofilms on denture surfaces.
Collapse
|
490
|
Li L, Wu H, Zhu S, Ji Z, Chi X, Xie F, Hao Y, Lu H, Yang F, Yan L, Zhang D, Jiang Y, Ni T. Discovery of Novel 7-Hydroxy-5-oxo-4,5-dihydrothieno[3,2- b]pyridine-6-carboxamide Derivatives with Potent and Selective Antifungal Activity against Cryptococcus Species. J Med Chem 2022; 65:11257-11269. [PMID: 35922963 DOI: 10.1021/acs.jmedchem.2c00794] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Cryptococcus neoformans and Cryptococcus gattii can cause fatal invasive infections, especially in immunocompromised patients. However, few antifungal drugs are available to help treat cryptococcosis. In this study, by compound library screening, we presented the first report of hit compound P163-0892, which had potent in vitro and in vivo antifungal activity against Cryptococcus spp. In vitro tests showed that P163-0892 was not cytotoxic and had highly selective and strong antifungal activities against Cryptococcus spp. with MIC values less than 1 μg/mL. Synergism of P163-0892 and fluconazole was also observed in vitro. The in vivo antifungal efficacy of P163-0892 was assessed in a wax moth larval fungal infection model, and treatment with 10 mg/kg P163-0892 caused a significant reduction in fungal burden and significant extension of the survival time. Taken together, our data indicate that the hit compound P163-0892 warrants further investigation as a novel anti-Cryptococcus agent.
Collapse
Affiliation(s)
- Liping Li
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, No. 1239 Siping Road, Shanghai 200092, China
| | - Hao Wu
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, No. 1239 Siping Road, Shanghai 200092, China
| | - Shuo Zhu
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, No. 1239 Siping Road, Shanghai 200092, China
| | - Zhe Ji
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, No. 1239 Siping Road, Shanghai 200092, China
| | - Xiaochen Chi
- Department of Organic Chemistry, School of Pharmacy, Navy Medical University, PLA, No. 325 Guohe Road, Shanghai 200433, China
- School of Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Fei Xie
- Department of Organic Chemistry, School of Pharmacy, Navy Medical University, PLA, No. 325 Guohe Road, Shanghai 200433, China
| | - Yumeng Hao
- Department of Organic Chemistry, School of Pharmacy, Navy Medical University, PLA, No. 325 Guohe Road, Shanghai 200433, China
| | - Hui Lu
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, No. 1239 Siping Road, Shanghai 200092, China
| | - Feng Yang
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, No. 1239 Siping Road, Shanghai 200092, China
| | - Lan Yan
- Center for New Drug Research, School of Pharmacy, Navy Medical University, PLA, No. 325 Guohe Road, Shanghai 200433, China
| | - Dazhi Zhang
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, No. 1239 Siping Road, Shanghai 200092, China
- Department of Organic Chemistry, School of Pharmacy, Navy Medical University, PLA, No. 325 Guohe Road, Shanghai 200433, China
| | - Yuanying Jiang
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, No. 1239 Siping Road, Shanghai 200092, China
| | - Tingjunhong Ni
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, No. 1239 Siping Road, Shanghai 200092, China
| |
Collapse
|
491
|
Branco J, Ryan AP, Silva AP, Butler G, Miranda IM, Rodrigues AG. Clinical azole cross-resistance in Candida parapsilosis is related to a novel MRR1 gain-of-function mutation. Clin Microbiol Infect 2022; 28:1655.e5-1655.e8. [PMID: 36028086 DOI: 10.1016/j.cmi.2022.08.014] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 08/12/2022] [Accepted: 08/16/2022] [Indexed: 11/03/2022]
Abstract
OBJECTIVES Hereby is described the molecular mechanisms underlying the acquisition of azole resistance by a C. parapsilosis isolate following fluconazole treatment due to candiduria. METHODS A set of three consecutive C. parapsilosis isolates were recovered from urine samples of a patient with candiduria. Whole-genome sequencing (WGS) and antifungal susceptibility assays were performed. Expression of MRR1, MDR1, ERG11 and CDR1B (CPAR2_304370) was quantified by RT-qPCR. RESULTS The initial isolate CPS-A, was susceptible to all three azoles tested (fluconazole, voriconazole and posaconazole); isolate CPS-B, collected after the 2nd cycle of treatment, exhibited a susceptible-dose dependent phenotype to fluconazole, while isolate CPS-C, recovered after the 3rd cycle, exhibited a cross-resistance profile to fluconazole and voriconazole. WGS revealed a putative resistance mechanism in isolate CPS-C, associated with a G1810A nucleotide substitution, leading to a G604R change in the Mrr1p transcription factor. Introducing this mutation into the susceptible CPS-A isolate (MRR1RI) resulted in resistance to fluconazole and voriconazole, as well as upregulation of MRR1 and MDR1. Interestingly, the susceptible-dose dependent phenotype exhibited by isolate CPS-B is associated with an increased copy number of the CDR1B gene. Expression of CDR1B is increased in both isolates CPS-B and CPS-C, and in the MRR1RI strain, harboring the gain-of-function (GOF) mutation. CONCLUSIONS Our results describe clinical azole cross-resistance acquisition in C. parapsilosis due to a G1810A (G604R) GOF mutation resulting in MRR1 hyperactivation and consequently, MDR1 efflux pump overexpression. We also associated amplification of CDR1B gene with decreased fluconazole susceptibility and showed that it is a putative target of the MRR1 GOF mutation.
Collapse
Affiliation(s)
- Joana Branco
- Division of Microbiology, Department of Pathology, Faculty of Medicine, University of Porto, Porto, Portugal; Center for Health Technology and Services Research - CINTESIS@RISE, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Adam P Ryan
- School of Biomolecular and Biomedical Science, Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
| | - Ana Pinto Silva
- Division of Microbiology, Department of Pathology, Faculty of Medicine, University of Porto, Porto, Portugal; Center for Health Technology and Services Research - CINTESIS@RISE, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Geraldine Butler
- School of Biomolecular and Biomedical Science, Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
| | - Isabel M Miranda
- Cardiovascular Research & Development Centre - UnIC@RISE, Faculty of Medicine, University of Porto, Porto, Portugal.
| | - Acácio Gonçalves Rodrigues
- Division of Microbiology, Department of Pathology, Faculty of Medicine, University of Porto, Porto, Portugal; Center for Health Technology and Services Research - CINTESIS@RISE, Faculty of Medicine, University of Porto, Porto, Portugal
| |
Collapse
|
492
|
Epigenetic Regulation of Antifungal Drug Resistance. J Fungi (Basel) 2022; 8:jof8080875. [PMID: 36012862 PMCID: PMC9409733 DOI: 10.3390/jof8080875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 08/02/2022] [Accepted: 08/04/2022] [Indexed: 11/23/2022] Open
Abstract
In medical mycology, epigenetic mechanisms are emerging as key regulators of multiple aspects of fungal biology ranging from development, phenotypic and morphological plasticity to antifungal drug resistance. Emerging resistance to the limited therapeutic options for the treatment of invasive fungal infections is a growing concern. Human fungal pathogens develop drug resistance via multiple mechanisms, with recent studies highlighting the role of epigenetic changes involving the acetylation and methylation of histones, remodeling of chromatin and heterochromatin-based gene silencing, in the acquisition of antifungal resistance. A comprehensive understanding of how pathogens acquire drug resistance will aid the development of new antifungal therapies as well as increase the efficacy of current antifungals by blocking common drug-resistance mechanisms. In this article, we describe the epigenetic mechanisms that affect resistance towards widely used systemic antifungal drugs: azoles, echinocandins and polyenes. Additionally, we review the literature on the possible links between DNA mismatch repair, gene silencing and drug-resistance mechanisms.
Collapse
|
493
|
Invasive Fungal Infection Caused by Magnusiomyces capitatus in an Immunocompromised Pediatric Patient with Acute Lymphoblastic Leukemia in Mexico City: A Case Report. J Fungi (Basel) 2022; 8:jof8080851. [PMID: 36012839 PMCID: PMC9410127 DOI: 10.3390/jof8080851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 08/05/2022] [Accepted: 08/12/2022] [Indexed: 11/17/2022] Open
Abstract
Magnusiomyces capitatus (also denominated “Geotrichum capitatum” and “the teleomorph stage of Saprochaete capitata”) mainly affects immunocompromised patients with hematological malignancies in rare cases of invasive fungal infections (IFIs). Few cases have been reported for pediatric patients with acute lymphoblastic leukemia (ALL), in part because conventional diagnostic methods do not consistently detect M. capitatus in infections. The current contribution describes a systemic infection in a 15-year-old female diagnosed with ALL. She arrived at the Children’s Hospital of Mexico City with a fever and neutropenia and developed symptoms of septic shock 4 days later. M. capitatus ENCB-HI-834, the causal agent, was isolated from the patient’s blood, urine, bile, and peritoneal fluid samples. It was identified with matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOF MS) and a phylogenetic reconstruction using internal transcribed spacer (ITS) and 28S ribosomal sequences. The phylogenetic sequence of M. capitatus ENCB-HI-834 clustered with other M. capitatus-type strains with a 100% identity. In vitro antifungal testing, conducted with the Sensititre YeastOne susceptibility system, found the following minimum inhibitory concentration (MIC) values (μg/mL): posaconazole 0.25, amphotericin B 1.0, fluconazole > 8.0, itraconazole 0.25, ketoconazole 0.5, 5-flucytosine ≤ 0.06, voriconazole 0.25, and caspofungin > 16.0. No clinical breakpoints have been defined for M. capitatus. This is the first clinical case reported in Mexico of an IFI caused by M. capitatus in a pediatric patient with ALL. It emphasizes the importance of close monitoring for a timely and accurate diagnosis of neutropenia-related IFIs to determine the proper treatment with antibiotics, antifungals, and chemotherapy for instance including children with ALL.
Collapse
|
494
|
Aguiar TKB, Neto NAS, Freitas CDT, Silva AFB, Bezerra LP, Malveira EA, Branco LAC, Mesquita FP, Goldman GH, Alencar LMR, Oliveira JTA, Santos-Oliveira R, Souza PFN. Antifungal Potential of Synthetic Peptides against Cryptococcus neoformans: Mechanism of Action Studies Reveal Synthetic Peptides Induce Membrane-Pore Formation, DNA Degradation, and Apoptosis. Pharmaceutics 2022; 14:pharmaceutics14081678. [PMID: 36015304 PMCID: PMC9416200 DOI: 10.3390/pharmaceutics14081678] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 08/10/2022] [Accepted: 08/10/2022] [Indexed: 12/02/2022] Open
Abstract
Cryptococcus neoformans is a human-pathogenic yeast responsible for pneumonia and meningitis, mainly in patients immunocompromised. Infections caused by C. neoformans are a global health concern. Synthetic antimicrobial peptides (SAMPs) have emerged as alternative molecules to cope with fungal infections, including C. neoformans. Here, eight SAMPs were tested regarding their antifungal potential against C. neoformans and had their mechanisms of action elucidated by fluorescence and scanning electron microscopies. Five SAMPs showed an inhibitory effect (MIC50) on C. neoformans growth at low concentrations. Fluorescence microscope (FM) revealed that SAMPs induced 6-kDa pores in the C. neoformans membrane. Inhibitory assays in the presence of ergosterol revealed that some peptides lost their activity, suggesting interaction with it. Furthermore, FM analysis revealed that SAMPs induced caspase 3/7-mediated apoptosis and DNA degradation in C. neoformans cells. Scanning Electron Microscopy (SEM) analysis revealed that peptides induced many morphological alterations such as cell membrane, wall damage, and loss of internal content on C. neoformans cells. Our results strongly suggest synthetic peptides are potential alternative molecules to control C. neoformans growth and treat the cryptococcal infection.
Collapse
Affiliation(s)
- Tawanny K. B. Aguiar
- Department of Biochemistry and Molecular Biology, Federal University of Ceará, Fortaleza 60451-970, CE, Brazil
| | - Nilton A. S. Neto
- Department of Biochemistry and Molecular Biology, Federal University of Ceará, Fortaleza 60451-970, CE, Brazil
| | - Cleverson D. T. Freitas
- Department of Biochemistry and Molecular Biology, Federal University of Ceará, Fortaleza 60451-970, CE, Brazil
| | - Ayrles F. B. Silva
- Department of Biochemistry and Molecular Biology, Federal University of Ceará, Fortaleza 60451-970, CE, Brazil
| | - Leandro P. Bezerra
- Department of Fisheries Engineering, Federal University of Ceará, Fortaleza 60455-970, CE, Brazil
| | - Ellen A. Malveira
- Department of Biochemistry and Molecular Biology, Federal University of Ceará, Fortaleza 60451-970, CE, Brazil
| | - Levi A. C. Branco
- Department of Biochemistry and Molecular Biology, Federal University of Ceará, Fortaleza 60451-970, CE, Brazil
| | - Felipe P. Mesquita
- Drug Research and Development Center, Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza 60430-275, CE, Brazil
| | - Gustavo H. Goldman
- Faculty of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, São Paulo 14040-903, SP, Brazil
| | - Luciana M. R. Alencar
- Department of Physics, Laboratory of Biophysics and Nanosystems, Federal University of Maranhão, São Luís 65080-805, MA, Brazil
| | - Jose T. A. Oliveira
- Department of Biochemistry and Molecular Biology, Federal University of Ceará, Fortaleza 60451-970, CE, Brazil
| | - Ralph Santos-Oliveira
- Laboratory of Nanoradiopharmaceuticals and Radiopharmacy, Zona Oeste State University, Rio de Janeiro 23070-200, RJ, Brazil
- Brazilian Nuclear Energy Commission, Nuclear Engineering Institute, Rio de Janeiro 21941-906, RJ, Brazil
| | - Pedro F. N. Souza
- Department of Biochemistry and Molecular Biology, Federal University of Ceará, Fortaleza 60451-970, CE, Brazil
- Drug Research and Development Center, Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza 60430-275, CE, Brazil
- Correspondence:
| |
Collapse
|
495
|
Cai L, Gao P, Wang Z, Dai C, Ning Y, Ilkit M, Xue X, Xiao J, Chen C. Lung and gut microbiomes in pulmonary aspergillosis: Exploring adjunctive therapies to combat the disease. Front Immunol 2022; 13:988708. [PMID: 36032147 PMCID: PMC9411651 DOI: 10.3389/fimmu.2022.988708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 07/25/2022] [Indexed: 11/13/2022] Open
Abstract
Species within the Aspergillus spp. cause a wide range of infections in humans, including invasive pulmonary aspergillosis, chronic pulmonary aspergillosis, and allergic bronchopulmonary aspergillosis, and are associated with high mortality rates. The incidence of pulmonary aspergillosis (PA) is on the rise, and the emergence of triazole-resistant Aspergillus spp. isolates, especially Aspergillus fumigatus, limits the efficacy of mold-active triazoles. Therefore, host-directed and novel adjunctive therapies are required to more effectively combat PA. In this review, we focus on PA from a microbiome perspective. We provide a general overview of the effects of the lung and gut microbiomes on the growth of Aspergillus spp. and host immunity. We highlight the potential of the microbiome as a therapeutic target for PA.
Collapse
Affiliation(s)
- Liuyang Cai
- Shanghai Engineering Research Center of Lung Transplantation, Shanghai, China
- Basic School of Medicine, Second Military Medical University (Naval Medical University), Shanghai, China
| | - Peigen Gao
- Shanghai Engineering Research Center of Lung Transplantation, Shanghai, China
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Zeyu Wang
- Shanghai Engineering Research Center of Lung Transplantation, Shanghai, China
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Chenyang Dai
- Shanghai Engineering Research Center of Lung Transplantation, Shanghai, China
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Ye Ning
- Shanghai Engineering Research Center of Lung Transplantation, Shanghai, China
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Macit Ilkit
- Division of Mycology, Department of Microbiology, Faculty of Medicine, University of Çukurova, Adana, Turkey
| | - Xiaochun Xue
- Department of Pharmacy, 905th Hospital of People’s Liberation Army of China (PLA) Navy, Shanghai, China
- *Correspondence: Xiaochun Xue, ; Jinzhou Xiao, ; Chang Chen,
| | - Jinzhou Xiao
- Shanghai Engineering Research Center of Hadal Science and Technology, College of Marine Sciences, Shanghai Ocean University, Shanghai, China
- *Correspondence: Xiaochun Xue, ; Jinzhou Xiao, ; Chang Chen,
| | - Chang Chen
- Shanghai Engineering Research Center of Lung Transplantation, Shanghai, China
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
- *Correspondence: Xiaochun Xue, ; Jinzhou Xiao, ; Chang Chen,
| |
Collapse
|
496
|
Blackman LD, Sutherland TD, De Barro PJ, Thissen H, Locock KES. Addressing a future pandemic: how can non-biological complex drugs prepare us for antimicrobial resistance threats? MATERIALS HORIZONS 2022; 9:2076-2096. [PMID: 35703580 DOI: 10.1039/d2mh00254j] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Loss of effective antibiotics through antimicrobial resistance (AMR) is one of the greatest threats to human health. By 2050, the annual death rate resulting from AMR infections is predicted to have climbed from 1.27 million per annum in 2019, up to 10 million per annum. It is therefore imperative to preserve the effectiveness of both existing and future antibiotics, such that they continue to save lives. One way to conserve the use of existing antibiotics and build further contingency against resistant strains is to develop alternatives. Non-biological complex drugs (NBCDs) are an emerging class of therapeutics that show multi-mechanistic antimicrobial activity and hold great promise as next generation antimicrobial agents. We critically outline the focal advancements for each key material class, including antimicrobial polymer materials, carbon nanomaterials, and inorganic nanomaterials, and highlight the potential for the development of antimicrobial resistance against each class. Finally, we outline remaining challenges for their clinical translation, including the need for specific regulatory pathways to be established in order to allow for more efficient clinical approval and adoption of these new technologies.
Collapse
Affiliation(s)
- Lewis D Blackman
- CSIRO Manufacturing, Research Way, Clayton, VIC 3168, Australia.
| | - Tara D Sutherland
- CSIRO Health & Biosecurity, Clunies Ross Street, Black Mountain, ACT 2601, Australia
| | - Paul J De Barro
- CSIRO Health & Biosecurity, Boggo Road, Dutton Park, QLD 4102, Australia
| | - Helmut Thissen
- CSIRO Manufacturing, Research Way, Clayton, VIC 3168, Australia.
| | | |
Collapse
|
497
|
Perspectives for Uses of Propolis in Therapy against Infectious Diseases. Molecules 2022; 27:molecules27144594. [PMID: 35889466 PMCID: PMC9320184 DOI: 10.3390/molecules27144594] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 07/06/2022] [Accepted: 07/06/2022] [Indexed: 01/22/2023] Open
Abstract
Propolis has gained wide popularity over the last decades in several parts of the world. In parallel, the literature about propolis composition and biological properties increased markedly. A great number of papers have demonstrated that propolis from different parts of the world is composed mainly of phenolic substances, frequently flavonoids, derived from plant resins. Propolis has a relevant role in increasing the social immunity of bee hives. Experimental evidence indicates that propolis and its components have activity against bacteria, fungi, and viruses. Mechanisms of action on bacteria, fungi, and viruses are known for several propolis components. Experiments have shown that propolis may act synergistically with antibiotics, antifungals, and antivirus drugs, permitting the administration of lower doses of drugs and higher antimicrobial effects. The current trend of growing resistance of microbial pathogens to the available drugs has encouraged the introduction of propolis in therapy against infectious diseases. Because propolis composition is widely variable, standardized propolis extracts have been produced. Successful clinical trials have included propolis extracts as medicine in dentistry and as an adjuvant in the treatment of patients against COVID-19. Present world health conditions encourage initiatives toward the spread of the niche of propolis, not only as traditional and alternative medicine but also as a relevant protagonist in anti-infectious therapy. Production of propolis and other apiary products is environmentally friendly and may contribute to alleviating the current crisis of the decline of bee populations. Propolis production has had social-economic relevance in Brazil, providing benefits to underprivileged people.
Collapse
|
498
|
Jampilek J. Novel avenues for identification of new antifungal drugs and current challenges. Expert Opin Drug Discov 2022; 17:949-968. [PMID: 35787715 DOI: 10.1080/17460441.2022.2097659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION : Some of otherwise useful fungi are pathogenic to humans, and unfortunately, the number of these pathogens is increasing. In addition to common skin infections, these opportunistic pathogens are able to cause severe, often incurable, systemic mycoses. AREAS COVERED : The number of antifungal drugs is limited, especially drugs that can be used for systemic administration, and resistance to these drugs is very common. This review summarizes various approaches to the discovery and development of new antifungal drugs, provides an overview of the most important molecules in terms of basic (laboratory) research and compounds currently in clinical trials, and focuses on drug repurposing strategy, while providing an overview of drugs of other indications that have been tested in vitro for their antifungal activity for possible expansion of antifungal drugs and/or support of existing antimycotics. EXPERT OPINION : Despite the limitations of the research of new antifungal drugs by pharmaceutical manufacturers, in addition to innovated molecules based on clinically used drugs, several completely new small entities with unique mechanisms of actions have been identified. The identification of new molecular targets that offer alternatives for the development of new unique selective antifungal highly effective agents has been an important outcome of repurposing of non-antifungal drugs to antifungal drug. Also, given the advances in monoclonal antibodies and their application to immunosuppressed patients, it may seem possible to predict a more optimistic future for antifungal therapy than has been the case in recent decades.
Collapse
Affiliation(s)
- Josef Jampilek
- Department of Analytical Chemistry, Faculty of Natural Sciences, Comenius University, Ilkovicova 6, 842 15 Bratislava, Slovakia.,Institute of Neuroimmunology, Slovak Academy of Sciences, Dubravska Cesta 9, 845 10 Bratislava, Slovakia
| |
Collapse
|
499
|
Silva SG, Paula P, da Silva JP, Mil-Homens D, Teixeira MC, Fialho AM, Costa R, Keller-Costa T. Insights into the Antimicrobial Activities and Metabolomes of Aquimarina ( Flavobacteriaceae, Bacteroidetes) Species from the Rare Marine Biosphere. Mar Drugs 2022; 20:423. [PMID: 35877716 PMCID: PMC9323603 DOI: 10.3390/md20070423] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 06/16/2022] [Accepted: 06/24/2022] [Indexed: 12/17/2022] Open
Abstract
Two novel natural products, the polyketide cuniculene and the peptide antibiotic aquimarin, were recently discovered from the marine bacterial genus Aquimarina. However, the diversity of the secondary metabolite biosynthetic gene clusters (SM-BGCs) in Aquimarina genomes indicates a far greater biosynthetic potential. In this study, nine representative Aquimarina strains were tested for antimicrobial activity against diverse human-pathogenic and marine microorganisms and subjected to metabolomic and genomic profiling. We found an inhibitory activity of most Aquimarina strains against Candida glabrata and marine Vibrio and Alphaproteobacteria species. Aquimarina sp. Aq135 and Aquimarina muelleri crude extracts showed particularly promising antimicrobial activities, amongst others against methicillin-resistant Staphylococcus aureus. The metabolomic and functional genomic profiles of Aquimarina spp. followed similar patterns and were shaped by phylogeny. SM-BGC and metabolomics networks suggest the presence of novel polyketides and peptides, including cyclic depsipeptide-related compounds. Moreover, exploration of the ‘Sponge Microbiome Project’ dataset revealed that Aquimarina spp. possess low-abundance distributions worldwide across multiple marine biotopes. Our study emphasizes the relevance of this member of the microbial rare biosphere as a promising source of novel natural products. We predict that future metabologenomics studies of Aquimarina species will expand the spectrum of known secondary metabolites and bioactivities from marine ecosystems.
Collapse
Affiliation(s)
- Sandra Godinho Silva
- IBB—Institute for Bioengineering and Biosciences and i4HB—Institute for Health and Bioeconomy, Instituto Superior Técnico, Av. Rovisco Pais, 1049-001 Lisbon, Portugal; (S.G.S.); (P.P.); (D.M.-H.); (M.C.T.); (A.M.F.)
- Bioengeneering Department, Instituto Superior Técnico, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
| | - Patrícia Paula
- IBB—Institute for Bioengineering and Biosciences and i4HB—Institute for Health and Bioeconomy, Instituto Superior Técnico, Av. Rovisco Pais, 1049-001 Lisbon, Portugal; (S.G.S.); (P.P.); (D.M.-H.); (M.C.T.); (A.M.F.)
- Bioengeneering Department, Instituto Superior Técnico, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
| | - José Paulo da Silva
- Centre of Marine Sciences, University of Algarve, Campus de Gambelas, 8005-139 Faro, Portugal;
| | - Dalila Mil-Homens
- IBB—Institute for Bioengineering and Biosciences and i4HB—Institute for Health and Bioeconomy, Instituto Superior Técnico, Av. Rovisco Pais, 1049-001 Lisbon, Portugal; (S.G.S.); (P.P.); (D.M.-H.); (M.C.T.); (A.M.F.)
- Bioengeneering Department, Instituto Superior Técnico, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
| | - Miguel Cacho Teixeira
- IBB—Institute for Bioengineering and Biosciences and i4HB—Institute for Health and Bioeconomy, Instituto Superior Técnico, Av. Rovisco Pais, 1049-001 Lisbon, Portugal; (S.G.S.); (P.P.); (D.M.-H.); (M.C.T.); (A.M.F.)
- Bioengeneering Department, Instituto Superior Técnico, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
| | - Arsénio Mendes Fialho
- IBB—Institute for Bioengineering and Biosciences and i4HB—Institute for Health and Bioeconomy, Instituto Superior Técnico, Av. Rovisco Pais, 1049-001 Lisbon, Portugal; (S.G.S.); (P.P.); (D.M.-H.); (M.C.T.); (A.M.F.)
- Bioengeneering Department, Instituto Superior Técnico, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
| | - Rodrigo Costa
- IBB—Institute for Bioengineering and Biosciences and i4HB—Institute for Health and Bioeconomy, Instituto Superior Técnico, Av. Rovisco Pais, 1049-001 Lisbon, Portugal; (S.G.S.); (P.P.); (D.M.-H.); (M.C.T.); (A.M.F.)
- Bioengeneering Department, Instituto Superior Técnico, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
- Centre of Marine Sciences, University of Algarve, Campus de Gambelas, 8005-139 Faro, Portugal;
| | - Tina Keller-Costa
- IBB—Institute for Bioengineering and Biosciences and i4HB—Institute for Health and Bioeconomy, Instituto Superior Técnico, Av. Rovisco Pais, 1049-001 Lisbon, Portugal; (S.G.S.); (P.P.); (D.M.-H.); (M.C.T.); (A.M.F.)
- Bioengeneering Department, Instituto Superior Técnico, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
| |
Collapse
|
500
|
Stevenson EM, Gaze WH, Gow NAR, Hart A, Schmidt W, Usher J, Warris A, Wilkinson H, Murray AK. Antifungal Exposure and Resistance Development: Defining Minimal Selective Antifungal Concentrations and Testing Methodologies. FRONTIERS IN FUNGAL BIOLOGY 2022; 3:918717. [PMID: 37746188 PMCID: PMC10512330 DOI: 10.3389/ffunb.2022.918717] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 05/16/2022] [Indexed: 09/26/2023]
Abstract
This scoping review aims to summarise the current understanding of selection for antifungal resistance (AFR) and to compare and contrast this with selection for antibacterial resistance, which has received more research attention. AFR is an emerging global threat to human health, associated with high mortality rates, absence of effective surveillance systems and with few alternative treatment options available. Clinical AFR is well documented, with additional settings increasingly being recognised to play a role in the evolution and spread of AFR. The environment, for example, harbours diverse fungal communities that are regularly exposed to antifungal micropollutants, potentially increasing AFR selection risk. The direct application of effect concentrations of azole fungicides to agricultural crops and the incomplete removal of pharmaceutical antifungals in wastewater treatment systems are of particular concern. Currently, environmental risk assessment (ERA) guidelines do not require assessment of antifungal agents in terms of their ability to drive AFR development, and there are no established experimental tools to determine antifungal selective concentrations. Without data to interpret the selective risk of antifungals, our ability to effectively inform safe environmental thresholds is severely limited. In this review, potential methods to generate antifungal selective concentration data are proposed, informed by approaches used to determine antibacterial minimal selective concentrations. Such data can be considered in the development of regulatory guidelines that aim to reduce selection for AFR.
Collapse
Affiliation(s)
- Emily M. Stevenson
- European Centre for Environment and Human Health, University of Exeter Medical School, Cornwall, United Kingdom
- Environment and Sustainability Institute, University of Exeter Medical School, Cornwall, United Kingdom
| | - William H. Gaze
- European Centre for Environment and Human Health, University of Exeter Medical School, Cornwall, United Kingdom
- Environment and Sustainability Institute, University of Exeter Medical School, Cornwall, United Kingdom
| | - Neil A. R. Gow
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, United Kingdom
| | - Alwyn Hart
- Chief Scientist’s Group, Environment Agency, Horizon House, Bristol, England, United Kingdom
| | - Wiebke Schmidt
- Chief Scientist’s Group, Environment Agency, Horizon House, Bristol, England, United Kingdom
| | - Jane Usher
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, United Kingdom
| | - Adilia Warris
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, United Kingdom
| | - Helen Wilkinson
- Chief Scientist’s Group, Environment Agency, Horizon House, Bristol, England, United Kingdom
| | - Aimee K. Murray
- European Centre for Environment and Human Health, University of Exeter Medical School, Cornwall, United Kingdom
- Environment and Sustainability Institute, University of Exeter Medical School, Cornwall, United Kingdom
| |
Collapse
|