1
|
Rajão A, Silva JPN, Almeida-Nunes DL, Rompante P, Rodrigues CF, Andrade JC. Limosilactobacillus reuteri AJCR4: A Potential Probiotic in the Fight Against Oral Candida spp. Biofilms. Int J Mol Sci 2025; 26:638. [PMID: 39859352 PMCID: PMC11766303 DOI: 10.3390/ijms26020638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 01/06/2025] [Accepted: 01/12/2025] [Indexed: 01/27/2025] Open
Abstract
Oral candidiasis is one of the most common infections in the immunocompromised. Biofilms of Candida species can make treatments difficult, leading to oral infection recurrence. This research aimed to isolate a Lactobacillus with anti-Candida effects from the oral cavity. An oral Lactobacillus was isolated in caries-free individuals. The best isolate was evaluated against Candida spp. planktonic and biofilm forms. The bacterial impacts on Candida biofilms' adhesion to acrylic discs were analyzed through an in vitro test. L. reuteri AJCR4 had the best anti-Candida activity in the preliminary screening. Results were promising in both planktonic and biofilms, particularly with C. albicans SC5314 and C. tropicalis ATCC750, where no viable cells were detected when using the cell-free supernatant (undiluted). In C. glabrata ATCC2001 and C. parapsilosis ATCC22019 biofilms, reductions of 3 Log10 and more than 2 Log10, respectively, were noted when using a cell suspension of L. reuteri ACJR4 (108 CFU/mL). On polymethyl methacrylate acrylic discs, the cell-free supernatant reduced Candida adhesion, resulting in no viable cell detection on the surface. In conclusion, L. reuteri AJCR4 demonstrated notable antifungal activity against Candida biofilms. This oral isolate and its postbiotic can be a potential alternative strategy to oral candidiasis, especially to treat recalcitrant infections.
Collapse
Affiliation(s)
- António Rajão
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, University Institute of Health Sciences—CESPU, 4585-116 Gandra, Portugal; (A.R.); (D.L.A.-N.); (J.C.A.)
- UCIBIO—Applied Molecular Biosciences Unit, Translational Toxicology Research Laboratory, University Institute of Health Sciences (1H-TOXRUN, IUCS-CESPU), 4585-116 Gandra, Portugal
| | - João P. N. Silva
- UNIPRO—Oral Pathology and Rehabilitation Research Unit, CESPU, CRL, 4585-116 Gandra, Portugal;
| | - Diana L. Almeida-Nunes
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, University Institute of Health Sciences—CESPU, 4585-116 Gandra, Portugal; (A.R.); (D.L.A.-N.); (J.C.A.)
- UCIBIO—Applied Molecular Biosciences Unit, Translational Toxicology Research Laboratory, University Institute of Health Sciences (1H-TOXRUN, IUCS-CESPU), 4585-116 Gandra, Portugal
- Differentiation and Cancer Group, Institute for Research and Innovation in Health (i3S) of the University of Porto, 4200-135 Porto, Portugal
| | - Paulo Rompante
- UNIPRO—Oral Pathology and Rehabilitation Research Unit, CESPU, CRL, 4585-116 Gandra, Portugal;
| | - Célia Fortuna Rodrigues
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, University Institute of Health Sciences—CESPU, 4585-116 Gandra, Portugal; (A.R.); (D.L.A.-N.); (J.C.A.)
- UCIBIO—Applied Molecular Biosciences Unit, Translational Toxicology Research Laboratory, University Institute of Health Sciences (1H-TOXRUN, IUCS-CESPU), 4585-116 Gandra, Portugal
- LEPABE—Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, 4200-465 Porto, Portugal
- ALiCE—Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, 4200-465 Porto, Portugal
| | - José Carlos Andrade
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, University Institute of Health Sciences—CESPU, 4585-116 Gandra, Portugal; (A.R.); (D.L.A.-N.); (J.C.A.)
- UCIBIO—Applied Molecular Biosciences Unit, Translational Toxicology Research Laboratory, University Institute of Health Sciences (1H-TOXRUN, IUCS-CESPU), 4585-116 Gandra, Portugal
| |
Collapse
|
2
|
Czajka KM, Venkataraman K, Brabant-Kirwan D, Santi SA, Verschoor C, Appanna VD, Singh R, Saunders DP, Tharmalingam S. Molecular Mechanisms Associated with Antifungal Resistance in Pathogenic Candida Species. Cells 2023; 12:2655. [PMID: 37998390 PMCID: PMC10670235 DOI: 10.3390/cells12222655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 11/14/2023] [Accepted: 11/17/2023] [Indexed: 11/25/2023] Open
Abstract
Candidiasis is a highly pervasive infection posing major health risks, especially for immunocompromised populations. Pathogenic Candida species have evolved intrinsic and acquired resistance to a variety of antifungal medications. The primary goal of this literature review is to summarize the molecular mechanisms associated with antifungal resistance in Candida species. Resistance can be conferred via gain-of-function mutations in target pathway genes or their transcriptional regulators. Therefore, an overview of the known gene mutations is presented for the following antifungals: azoles (fluconazole, voriconazole, posaconazole and itraconazole), echinocandins (caspofungin, anidulafungin and micafungin), polyenes (amphotericin B and nystatin) and 5-fluorocytosine (5-FC). The following mutation hot spots were identified: (1) ergosterol biosynthesis pathway mutations (ERG11 and UPC2), resulting in azole resistance; (2) overexpression of the efflux pumps, promoting azole resistance (transcription factor genes: tac1 and mrr1; transporter genes: CDR1, CDR2, MDR1, PDR16 and SNQ2); (3) cell wall biosynthesis mutations (FKS1, FKS2 and PDR1), conferring resistance to echinocandins; (4) mutations of nucleic acid synthesis/repair genes (FCY1, FCY2 and FUR1), resulting in 5-FC resistance; and (5) biofilm production, promoting general antifungal resistance. This review also provides a summary of standardized inhibitory breakpoints obtained from international guidelines for prominent Candida species. Notably, N. glabrata, P. kudriavzevii and C. auris demonstrate fluconazole resistance.
Collapse
Affiliation(s)
- Karolina M. Czajka
- Medical Sciences Division, NOSM University, 935 Ramsey Lake Rd., Sudbury, ON P3E 2C6, Canada; (K.M.C.); (K.V.); (C.V.); (R.S.); (D.P.S.)
| | - Krishnan Venkataraman
- Medical Sciences Division, NOSM University, 935 Ramsey Lake Rd., Sudbury, ON P3E 2C6, Canada; (K.M.C.); (K.V.); (C.V.); (R.S.); (D.P.S.)
- School of Natural Sciences, Laurentian University, Sudbury, ON P3E 2C6, Canada;
| | | | - Stacey A. Santi
- Health Sciences North Research Institute, Sudbury, ON P3E 2H2, Canada; (D.B.-K.); (S.A.S.)
| | - Chris Verschoor
- Medical Sciences Division, NOSM University, 935 Ramsey Lake Rd., Sudbury, ON P3E 2C6, Canada; (K.M.C.); (K.V.); (C.V.); (R.S.); (D.P.S.)
- School of Natural Sciences, Laurentian University, Sudbury, ON P3E 2C6, Canada;
- Health Sciences North Research Institute, Sudbury, ON P3E 2H2, Canada; (D.B.-K.); (S.A.S.)
| | - Vasu D. Appanna
- School of Natural Sciences, Laurentian University, Sudbury, ON P3E 2C6, Canada;
| | - Ravi Singh
- Medical Sciences Division, NOSM University, 935 Ramsey Lake Rd., Sudbury, ON P3E 2C6, Canada; (K.M.C.); (K.V.); (C.V.); (R.S.); (D.P.S.)
- Health Sciences North Research Institute, Sudbury, ON P3E 2H2, Canada; (D.B.-K.); (S.A.S.)
| | - Deborah P. Saunders
- Medical Sciences Division, NOSM University, 935 Ramsey Lake Rd., Sudbury, ON P3E 2C6, Canada; (K.M.C.); (K.V.); (C.V.); (R.S.); (D.P.S.)
- Health Sciences North Research Institute, Sudbury, ON P3E 2H2, Canada; (D.B.-K.); (S.A.S.)
| | - Sujeenthar Tharmalingam
- Medical Sciences Division, NOSM University, 935 Ramsey Lake Rd., Sudbury, ON P3E 2C6, Canada; (K.M.C.); (K.V.); (C.V.); (R.S.); (D.P.S.)
- School of Natural Sciences, Laurentian University, Sudbury, ON P3E 2C6, Canada;
- Health Sciences North Research Institute, Sudbury, ON P3E 2H2, Canada; (D.B.-K.); (S.A.S.)
| |
Collapse
|
3
|
Caballero U, Eraso E, Pemán J, Quindós G, Vozmediano V, Schmidt S, Jauregizar N. In Vitro Pharmacokinetic/Pharmacodynamic Modelling and Simulation of Amphotericin B against Candida auris. Pharmaceutics 2021; 13:pharmaceutics13111767. [PMID: 34834182 PMCID: PMC8624019 DOI: 10.3390/pharmaceutics13111767] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 10/15/2021] [Accepted: 10/18/2021] [Indexed: 11/25/2022] Open
Abstract
The aims of this study were to characterize the antifungal activity of amphotericin B against Candida auris in a static in vitro system and to evaluate different dosing schedules and MIC scenarios by means of semi-mechanistic pharmacokinetic/pharmacodynamic (PK/PD) modelling and simulation. A two-compartment model consisting of a drug-susceptible and a drug-resistant subpopulation successfully characterized the time-kill data and a modified Emax sigmoidal model best described the effect of the drug. The model incorporated growth rate constants for both subpopulations, a death rate constant and a transfer constant between both compartments. Additionally, the model included a parameter to account for the delay in growth in the absence or presence of the drug. Amphotericin B displayed a concentration-dependent fungicidal activity. The developed PK/PD model was able to characterize properly the antifungal activity of amphotericin B against C. auris. Finally, simulation analysis revealed that none of the simulated standard dosing scenarios of 0.6, 1 and 1.5 mg/kg/day over a week treatment showed successful activity against C. auris infection. Simulations also pointed out that an MIC of 1 mg/L would be linked to treatment failure for C. auris invasive infections and therefore, the resistance rate to amphotericin B may be higher than previously reported.
Collapse
Affiliation(s)
- Unai Caballero
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain;
| | - Elena Eraso
- Department of Immunology, Microbiology and Parasitology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain; (E.E.); (G.Q.)
| | - Javier Pemán
- Microbiology Department, Hospital Universitario y Politécnico de La Fe, 46026 Valencia, Spain;
- Severe Infection Research Group, Health Research Institute Hospital La Fe, 46026 Valencia, Spain
| | - Guillermo Quindós
- Department of Immunology, Microbiology and Parasitology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain; (E.E.); (G.Q.)
| | - Valvanera Vozmediano
- Center for Pharmacometrics and Systems Pharmacology, Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando, FL 32827, USA; (V.V.); (S.S.)
| | - Stephan Schmidt
- Center for Pharmacometrics and Systems Pharmacology, Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando, FL 32827, USA; (V.V.); (S.S.)
| | - Nerea Jauregizar
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain;
- Correspondence:
| |
Collapse
|
4
|
Garcia-Effron G. Rezafungin-Mechanisms of Action, Susceptibility and Resistance: Similarities and Differences with the Other Echinocandins. J Fungi (Basel) 2020; 6:E262. [PMID: 33139650 PMCID: PMC7711656 DOI: 10.3390/jof6040262] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 10/19/2020] [Accepted: 10/22/2020] [Indexed: 12/20/2022] Open
Abstract
Rezafungin (formerly CD101) is a new β-glucan synthase inhibitor that is chemically related with anidulafungin. It is considered the first molecule of the new generation of long-acting echinocandins. It has several advantages over the already approved by the Food and Drug Administration (FDA) echinocandins as it has better tissue penetration, better pharmacokinetic/phamacodynamic (PK/PD) pharmacometrics, and a good safety profile. It is much more stable in solution than the older echinocandins, making it more flexible in terms of dosing, storage, and manufacturing. These properties would allow rezafungin to be administered once-weekly (intravenous) and to be potentially administered topically and subcutaneously. In addition, higher dose regimens were tested with no evidence of toxic effect. This will eventually prevent (or reduce) the selection of resistant strains. Rezafungin also has several similarities with older echinocandins as they share the same in vitro behavior (very similar Minimum Inhibitory Concentration required to inhibit the growth of 50% of the isolates (MIC50) and half enzyme maximal inhibitory concentration 50% (IC50)) and spectrum, the same target, and the same mechanisms of resistance. The selection of FKS mutants occurred at similar frequency for rezafungin than for anidulafungin and caspofungin. In this review, rezafungin mechanism of action, target, mechanism of resistance, and in vitro data are described in a comparative manner with the already approved echinocandins.
Collapse
Affiliation(s)
- Guillermo Garcia-Effron
- Laboratorio de Micología y Diagnóstico Molecular, Cátedra de Parasitología y Micología, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, C.P. 3000 Santa Fe, Argentina; or ; Tel.: +54-9342-4575209 (ext. 135)
- Consejo Nacional de Investigaciones Científicas y Tecnológicas, C.P. 3000 Santa Fe, Argentina
| |
Collapse
|
5
|
Dudiuk C, Berrio I, Leonardelli F, Morales-Lopez S, Theill L, Macedo D, Yesid-Rodriguez J, Salcedo S, Marin A, Gamarra S, Garcia-Effron G. Antifungal activity and killing kinetics of anidulafungin, caspofungin and amphotericin B against Candida auris. J Antimicrob Chemother 2019; 74:2295-2302. [DOI: 10.1093/jac/dkz178] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Revised: 03/08/2019] [Accepted: 04/02/2019] [Indexed: 01/21/2023] Open
Abstract
AbstractBackgroundCandida auris is an emerging MDR pathogen. It shows reduced susceptibility to azole drugs and, in some strains, high amphotericin B MICs have been described. For these reasons, echinocandins were proposed as first-line treatment for C. auris infections. However, information on how echinocandins and amphotericin B act against this species is lacking.ObjectivesOur aim was to establish the killing kinetics of anidulafungin, caspofungin and amphotericin B against C. auris by time–kill methodology and to determine if these antifungals behave as fungicidal or fungistatic agents against this species.MethodsThe susceptibility of 50 C. auris strains was studied. Nine strains were selected (based on echinocandin MICs) to be further studied. Minimal fungicidal concentrations, in vitro dose–response and time–kill patterns were determined.ResultsEchinocandins showed lower MIC values than amphotericin B (geometric mean of 0.12 and 0.94 mg/L, respectively). Anidulafungin and caspofungin showed no fungicidal activity at any concentration (maximum log decreases in cfu/mL between 1.34 and 2.22). On the other hand, amphotericin B showed fungicidal activity, but at high concentrations (≥2.00 mg/L). In addition, the tested polyene was faster than echinocandins at killing 50% of the initial inoculum (0.92 versus >8.00 h, respectively).ConclusionsAmphotericin B was the only agent regarded as fungicidal against C. auris. Moreover, C. auris should be considered tolerant to caspofungin and anidulafungin considering that their MFC:MIC ratios were mostly ≥32 and that after 6 h of incubation the starting inoculum was not reduced in >90%.
Collapse
Affiliation(s)
- Catiana Dudiuk
- Laboratorio de Micología y Diagnóstico Molecular, Cátedra de Parasitología y Micología, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina
- Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), CCT, Santa Fe, Argentina
| | - Indira Berrio
- Medical and Experimental Mycology Group, Corporación para Investigaciones Biológicas (CIB), Medellín, Colombia
- Hospital general de Medellin ‘Luz Castro de Gutiérrez’ ESE, Medellín, Colombia
| | - Florencia Leonardelli
- Laboratorio de Micología y Diagnóstico Molecular, Cátedra de Parasitología y Micología, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina
- Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), CCT, Santa Fe, Argentina
| | - Soraya Morales-Lopez
- Universidad de Santander, Facultad de Ciencias de la Salud, Grupo de Investigación CIENCIA UDES, Valledupar, Colombia
- Laboratorios Nancy Flórez García S.A.S., Valledupar, Colombia
| | - Laura Theill
- Laboratorio de Micología y Diagnóstico Molecular, Cátedra de Parasitología y Micología, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina
| | - Daiana Macedo
- Laboratorio de Micología y Diagnóstico Molecular, Cátedra de Parasitología y Micología, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina
- Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), CCT, Santa Fe, Argentina
| | - José Yesid-Rodriguez
- Centro de Investigaciones Microbiológicas del Cesar, CIMCE, Valledupar, Colombia
| | | | | | - Soledad Gamarra
- Laboratorio de Micología y Diagnóstico Molecular, Cátedra de Parasitología y Micología, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina
| | - Guillermo Garcia-Effron
- Laboratorio de Micología y Diagnóstico Molecular, Cátedra de Parasitología y Micología, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina
- Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), CCT, Santa Fe, Argentina
| |
Collapse
|
6
|
Vieira JN, Feijó AM, Bueno ME, Gonçalves CL, Lund RG, Mendes JF, Villarreal JPV, Villela MM, Nascente PS. Evaluation of the frequency of Candida spp. in hospitalized and non-hospitalized subjects. BRAZ J BIOL 2018; 78:644-652. [PMID: 29451605 DOI: 10.1590/1519-6984.169623] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 05/27/2017] [Indexed: 11/21/2022] Open
Abstract
The aim of this study was to evaluate the frequency of Candida species between a non-hospitalized and a hospitalized population. For this purpose, samples of saliva were sampled through sterile swabs, moistened in peptone water and rubbed in the oral cavity of 140 individuals, from which, 70 were hospitalized patients from the Medical Clinic of a Teaching Hospital and the other 70 were non-hospitalized subjects. All saliva samples were plated in Sabouraud Dextrose agar added with Chloramphenicol and incubated at 36 °C for 48 hours. The morphology identification was performed through macroscopic and microscopic characterization, the CHROMagar Candida medium and the VITEK® system Yeast Biochemical Card (bio Mérieux SA, France). The results showed a colonization of Candida spp. in 85.7% the hospitalized individuals, where the species found were C. albicans (60%), C. tropicalis (23.4%), C. krusei (3.3%) and Candida spp. (13.3%). In the non-hospitalized individuals the colonization by Candida spp was 47.1%, and the species found were: C. albicans (45.5%), C.krusei (9.1%), C. guilliermondii (9.1% %), C. tropicalis (3.0%), C. famata (3.0%) and Candida spp. (30.3%). In spite of their presence in oral cavity in both groups, Candida spp. was more frequently isolated in hospitalized individuals, who were 6.73 times more likely to have this fungus in the oral cavity and were 3.88 times more likely to have Candida albicans.
Collapse
Affiliation(s)
- J N Vieira
- Laboratório de Parasitologia, Instituto de Biologia, Departamento de Microbiologia e Parasitologia, Universidade Federal de Pelotas - UFPEL, Campus universitário Capão do Leão, s/n, Prédio 18, Sala 3, CEP 96010-900, Pelotas, RS, Brazil
| | - A M Feijó
- Laboratório de Parasitologia, Instituto de Biologia, Departamento de Microbiologia e Parasitologia, Universidade Federal de Pelotas - UFPEL, Campus universitário Capão do Leão, s/n, Prédio 18, Sala 3, CEP 96010-900, Pelotas, RS, Brazil
| | - M E Bueno
- Laboratório de Parasitologia, Instituto de Biologia, Departamento de Microbiologia e Parasitologia, Universidade Federal de Pelotas - UFPEL, Campus universitário Capão do Leão, s/n, Prédio 18, Sala 3, CEP 96010-900, Pelotas, RS, Brazil
| | - C L Gonçalves
- Laboratório de Parasitologia, Instituto de Biologia, Departamento de Microbiologia e Parasitologia, Universidade Federal de Pelotas - UFPEL, Campus universitário Capão do Leão, s/n, Prédio 18, Sala 3, CEP 96010-900, Pelotas, RS, Brazil
| | - R G Lund
- Departamento de Odontologia Restauradora, Faculdade de Medicina Dentária, Universidade Federal de Pelotas - UFPEL, Rua Gonçalves Chaves, nº 457, Centro, CEP 96020-080, Pelotas, RS, Brazil
| | - J F Mendes
- Laboratório de Parasitologia, Instituto de Biologia, Departamento de Microbiologia e Parasitologia, Universidade Federal de Pelotas - UFPEL, Campus universitário Capão do Leão, s/n, Prédio 18, Sala 3, CEP 96010-900, Pelotas, RS, Brazil
| | - J P V Villarreal
- Laboratório de Parasitologia, Instituto de Biologia, Departamento de Microbiologia e Parasitologia, Universidade Federal de Pelotas - UFPEL, Campus universitário Capão do Leão, s/n, Prédio 18, Sala 3, CEP 96010-900, Pelotas, RS, Brazil
| | - M M Villela
- Laboratório de Parasitologia, Instituto de Biologia, Departamento de Microbiologia e Parasitologia, Universidade Federal de Pelotas - UFPEL, Campus universitário Capão do Leão, s/n, Prédio 18, Sala 3, CEP 96010-900, Pelotas, RS, Brazil
| | - P S Nascente
- Laboratório de Parasitologia, Instituto de Biologia, Departamento de Microbiologia e Parasitologia, Universidade Federal de Pelotas - UFPEL, Campus universitário Capão do Leão, s/n, Prédio 18, Sala 3, CEP 96010-900, Pelotas, RS, Brazil
| |
Collapse
|
7
|
Sanchis M, Martin-Vicente A, Capilla J, Guarro J. Antifungal therapies in murine infections byCandida kefyr. Mycoses 2016; 59:253-258. [DOI: 10.1111/myc.12468] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Revised: 12/21/2015] [Accepted: 12/22/2015] [Indexed: 11/28/2022]
Affiliation(s)
- Marta Sanchis
- Unitat de Microbiologia; Facultat de Medicina i Ciències de la Salut; IISPV; Universitat Rovira i Virgili; Reus Tarragona Spain
| | - Adela Martin-Vicente
- Unitat de Microbiologia; Facultat de Medicina i Ciències de la Salut; IISPV; Universitat Rovira i Virgili; Reus Tarragona Spain
| | - Javier Capilla
- Unitat de Microbiologia; Facultat de Medicina i Ciències de la Salut; IISPV; Universitat Rovira i Virgili; Reus Tarragona Spain
| | - Josep Guarro
- Unitat de Microbiologia; Facultat de Medicina i Ciències de la Salut; IISPV; Universitat Rovira i Virgili; Reus Tarragona Spain
| |
Collapse
|
8
|
Mazari W, Boucherit-Otmani Z, Boucherit K. In vitro susceptibility of amphotericin-B, voriconazole and caspofungin against Candida guilliermondii biofilms, isolated from dentals units water pipes, under different growth phases. J Mycol Med 2015; 25:57-62. [DOI: 10.1016/j.mycmed.2014.10.027] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Revised: 10/01/2014] [Accepted: 10/30/2014] [Indexed: 11/29/2022]
|
9
|
Maubon D, Garnaud C, Calandra T, Sanglard D, Cornet M. Resistance of Candida spp. to antifungal drugs in the ICU: where are we now? Intensive Care Med 2014; 40:1241-55. [PMID: 25091787 DOI: 10.1007/s00134-014-3404-7] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Accepted: 07/10/2014] [Indexed: 12/11/2022]
Abstract
Current increases in antifungal drug resistance in Candida spp. and clinical treatment failures are of concern, as invasive candidiasis is a significant cause of mortality in intensive care units (ICUs). This trend reflects the large and expanding use of newer broad-spectrum antifungal agents, such as triazoles and echinocandins. In this review, we firstly present an overview of the mechanisms of action of the drugs and of resistance in pathogenic yeasts, subsequently focusing on recent changes in the epidemiology of antifungal resistance in ICU. Then, we emphasize the clinical impacts of these current trends. The emergence of clinical treatment failures due to resistant isolates is described. We also consider the clinical usefulness of recent advances in the interpretation of antifungal susceptibility testing and in molecular detection of the mutations underlying acquired resistance. We pay particular attention to practical issues relating to ICU patient management, taking into account the growing threat of antifungal drug resistance.
Collapse
Affiliation(s)
- Danièle Maubon
- Parasitologie-Mycologie, Institut de Biologie et de Pathologie, CHU de Grenoble, Grenoble, France,
| | | | | | | | | |
Collapse
|
10
|
Paredes K, Pastor FJ, Capilla J, Sutton DA, Mayayo E, Fothergill AW, Guarro J. Therapies against murine Candida guilliermondii infection, relationship between in vitro antifungal pharmacodynamics and outcome. Rev Iberoam Micol 2014; 32:34-9. [PMID: 24594291 DOI: 10.1016/j.riam.2013.10.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2013] [Revised: 10/09/2013] [Accepted: 10/11/2013] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Candida guilliermondii has been recognized as an emerging pathogen showing a decreased susceptibility to fluconazole and considerably high echinocandin MICs. AIMS Evaluate the in vitro activity of anidulafungin in comparison to amphotericin B and fluconazole against different isolates of C. guilliermondii, and their efficacy in an immunosuppressed murine model of disseminated infection. METHODS The in vitro susceptibility of four strains against amphotericin B, fluconazole and anidulafungin was performed by using a reference broth microdilution method and time-kill curves. The in vivo efficacy was evaluated by determination of fungal load reduction in kidneys of infected animals receiving deoxycholate AMB at 0,8 mg/kg i.v., liposomal amphotericin B at 10 mg/kg i.v., fluconazole at 50 mg/kg, or anidulafungin at 10 mg/kg. RESULTS Amphotericin B and anidulafungin showed fungicidal activity, while fluconazole was fungistatic for all the strains. In the murine model, liposomal amphotericin B at 10 mg/kg/day was effective in reducing the tissue burden in kidneys of mice infected with any of the tested strains. However, amphotericin B, anidulafungin and fluconazole were only effective against those strains showing low MIC values. CONCLUSIONS Liposomal amphotericin B showed the higher activity and efficacy against the two strains of C. guilliermondii, in contrast to the poor effect of fluconazole and anidulafungin. Further studies with more isolates of C. guilliermondii representing a wider range of MICs should be carried out to assess whether there is any relationship between MIC values and anidulafungin efficacy.
Collapse
Affiliation(s)
- Katihuska Paredes
- Unitat de Microbiologia, Facultat de Medicina i Ciències de la Salut, IISPV, Universitat Rovira i Virgili, Reus, Tarragona, Spain
| | - Francisco Javier Pastor
- Unitat de Microbiologia, Facultat de Medicina i Ciències de la Salut, IISPV, Universitat Rovira i Virgili, Reus, Tarragona, Spain
| | - Javier Capilla
- Unitat de Microbiologia, Facultat de Medicina i Ciències de la Salut, IISPV, Universitat Rovira i Virgili, Reus, Tarragona, Spain.
| | - Deanna A Sutton
- Fungus Testing Laboratory, University of Texas Health Science Center, San Antonio, TX, USA
| | - Emilio Mayayo
- Unitat de Anatomia Patològica, Facultat de Medicina i Ciències de la Salut, IISPV, Universitat Rovira i Virgili, Reus, Tarragona, Spain
| | - Annette W Fothergill
- Fungus Testing Laboratory, University of Texas Health Science Center, San Antonio, TX, USA
| | - Josep Guarro
- Unitat de Microbiologia, Facultat de Medicina i Ciències de la Salut, IISPV, Universitat Rovira i Virgili, Reus, Tarragona, Spain
| |
Collapse
|
11
|
Spampinato C, Leonardi D. Candida infections, causes, targets, and resistance mechanisms: traditional and alternative antifungal agents. BIOMED RESEARCH INTERNATIONAL 2013; 2013:204237. [PMID: 23878798 PMCID: PMC3708393 DOI: 10.1155/2013/204237] [Citation(s) in RCA: 219] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/08/2013] [Revised: 06/06/2013] [Accepted: 06/06/2013] [Indexed: 11/25/2022]
Abstract
The genus Candida includes about 200 different species, but only a few species are human opportunistic pathogens and cause infections when the host becomes debilitated or immunocompromised. Candida infections can be superficial or invasive. Superficial infections often affect the skin or mucous membranes and can be treated successfully with topical antifungal drugs. However, invasive fungal infections are often life-threatening, probably due to inefficient diagnostic methods and inappropriate initial antifungal therapies. Here, we briefly review our current knowledge of pathogenic species of the genus Candida and yeast infection causes and then focus on current antifungal drugs and resistance mechanisms. An overview of new therapeutic alternatives for the treatment of Candida infections is also provided.
Collapse
Affiliation(s)
- Claudia Spampinato
- Departamento de Química Biológica, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario (UNR), Suipacha 531, 2000 Rosario, Argentina
- Centro de Estudios Fotosintéticos y Bioquímicos (CEFOBI, UNR-CONICET), Suipacha 531, 2000 Rosario, Argentina
| | - Darío Leonardi
- Departamento de Tecnología Farmacéutica, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario (UNR), Suipacha 531, 2000 Rosario, Argentina
- Instituto de Química Rosario (IQUIR, UNR-CONICET), Suipacha 531, 2000 Rosario, Argentina
| |
Collapse
|
12
|
Cantón E, Pemán J, Hervás D, Espinel-Ingroff A. Examination of the in vitro fungicidal activity of echinocandins against Candida lusitaniae by time-killing methods. J Antimicrob Chemother 2012; 68:864-8. [PMID: 23228935 DOI: 10.1093/jac/dks489] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
OBJECTIVES Candida lusitaniae fungaemia, although infrequent (1%), is more common in immunocompromised patients than Candida albicans. Although infections produced by Candida spp. are therapeutic targets for treatment with echinocandins, little information is available regarding their killing kinetics against C. lusitaniae. The objectives of this study were to determine the killing kinetics of anidulafungin, micafungin and caspofungin against four blood isolates of C. lusitaniae by time-kill methodology. METHODS Time-kill studies were performed in RMPI 1640 medium (5 mL, inoculum ∼10(5) cfu/mL). The number of cfu/mL was determined at 0, 2, 4, 6 and 24 h. The anidulafungin concentrations assayed were 0.03, 0.12, 0.5, 2 and 8 mg/L, while micafungin and caspofungin concentrations were 0.25, 1, 4, 16 and 32 mg/L. RESULTS MIC ranges were 0.03-1 mg/L (anidulafungin), 0.016-0.06 mg/L (micafungin) and 0.03-1 mg/L (caspofungin). The mean maximum log decrease in cfu/mL was reached with 2 mg/L anidulafungin (1.85 ± 0.4 log), 32 mg/L caspofungin (5.5 ± 0.2 log) and 32 mg/L micafungin (2.65 ± 1.9 log). Only caspofungin and micafungin reached the fungicidal endpoint (99.9% growth reduction or a 3 log decrease) with 32 mg/L at 22.8 h (caspofungin) and 26.5 h (micafungin). Analysis of variance showed significant differences in killing activity among isolates, but not among concentrations reached in serum or echinocandins. CONCLUSIONS Anidulafungin and micafungin exhibit greater killing rates than caspofungin. Caspofungin was the only echinocandin that reached the fungicidal endpoint before 24 h, but at drug concentrations (≥ 16 mg/L) not usually reached in serum. The echinocandin killing rate was isolate dependent and concentration independent.
Collapse
Affiliation(s)
- Emilia Cantón
- Unidad de Microbiología Experimental, Centro de Investigación, Hospital Universitario y Politécnico La Fe, Valencia 46009, Spain.
| | | | | | | |
Collapse
|
13
|
Elevated chitin content reduces the susceptibility of Candida species to caspofungin. Antimicrob Agents Chemother 2012; 57:146-54. [PMID: 23089748 DOI: 10.1128/aac.01486-12] [Citation(s) in RCA: 153] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The echinocandin antifungal drugs inhibit synthesis of the major fungal cell wall polysaccharide β(1,3)-glucan. Echinocandins have good efficacy against Candida albicans but reduced activity against other Candida species, in particular Candida parapsilosis and Candida guilliermondii. Treatment of Candida albicans with a sub-MIC level of caspofungin has been reported to cause a compensatory increase in chitin content and to select for sporadic echinocandin-resistant FKS1 point mutants that also have elevated cell wall chitin. Here we show that elevated chitin in response to caspofungin is a common response in various Candida species. Activation of chitin synthesis was observed in isolates of C. albicans, Candida tropicalis, C. parapsilosis, and C. guilliermondii and in some isolates of Candida krusei in response to caspofungin treatment. However, Candida glabrata isolates demonstrated no exposure-induced change in chitin content. Furthermore, isolates of C. albicans, C. krusei, C. parapsilosis, and C. guilliermondii which were stimulated to have higher chitin levels via activation of the calcineurin and protein kinase C (PKC) signaling pathways had reduced susceptibility to caspofungin. Isolates containing point mutations in the FKS1 gene generally had higher chitin levels and did not demonstrate a further compensatory increase in chitin content in response to caspofungin treatment. These results highlight the potential of increased chitin synthesis as a potential mechanism of tolerance to caspofungin for the major pathogenic Candida species.
Collapse
|
14
|
Espinel-Ingroff A, Cantón E. In vitro activity of echinocandins against non-Candida albicans: is echinocandin antifungal activity the same? Enferm Infecc Microbiol Clin 2011; 29 Suppl 2:3-9. [PMID: 21420570 DOI: 10.1016/s0213-005x(11)70002-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The echinocandins anidulafungin, caspofungin, and micafungin have a broad and similar spectrum of in vitro and in vivo activity against most Candida spp. Minimal inhibitory concentrations (MICs) for Candida spp. are usually below 1 μg/mL for most isolates. The exceptions are Candidaparapsilosis and C. guilliermondii. Species-specific clinical breakpoints (CBPs) and epidemiologic cutoff values (ECVs) have been proposed by the Clinical and Laboratory Standards Institute (CLSI) for the eight most common Candida spp. versus each echinocandin; these values are useful to detect in vitro antifungal resistance (CBPs) and to identify isolates harboring fks mutations or having reduced susceptibility (ECVs). This paper presents a review of the literature (2006-2010) regarding the in vitro activity similarities or differences among the three echinocandins against Candida spp.; different parameters or measurements of in vitro potency were evaluated. The focus of the review is the non-Candida albicans species.
Collapse
|
15
|
Saribas Z, Yurdakul P, Cetin-Hazirolan G, Arikan-Akdagli S. Influence of serum on in vitro susceptibility testing of echinocandins for Candida parapsilosis and Candida guilliermondii. Mycoses 2011; 55:156-60. [PMID: 21668525 DOI: 10.1111/j.1439-0507.2011.02050.x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Echinocandins are antifungal drugs used for the treatment of invasive candidiasis and aspergillosis. They bind to serum proteins within a rate of 96 to >99%. The effect of serum on in vitro echinocandin susceptibility tests of certain Candida and Aspergillus species was reported. This study was performed to determine the effect of human serum on in vitro susceptibility testing of echinocandins for clinical isolates of Candida parapsilosis and Candida guilliermondii, the species which generally have higher minimum inhibitor concentrations compared with other Candida species. One hundred C. parapsilosis and 20 C. guilliermondii isolates were included in the study. The susceptibility tests of caspofungin, micafungin and anidulafungin were performed using microdilution method, either in the presence or absence of 50% human serum, according to the Clinical and Laboratory Standards Institute (CLSI) M27-A3 guidelines. It was demonstrated that human serum significantly affects the in vitro susceptibility results of echinocandins for C. parapsilosis and C. guilliermondii isolates, mostly yielding an increase in MICs. The most prominent fold changes were for micafungin and anidulafungin in C. parapsilosis, and for anidulafungin in C. guilliermondii isolates. Serum influences the in vitro echinocandin susceptibility in C. parapsilosis and C. guilliermondii. The mechanism and clinical significance of this in vitro change need to be clarified.
Collapse
Affiliation(s)
- Zeynep Saribas
- Department of Medical Microbiology, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | | | | | | |
Collapse
|
16
|
Benincasa M, Pacor S, Wu W, Prato M, Bianco A, Gennaro R. Antifungal activity of amphotericin B conjugated to carbon nanotubes. ACS NANO 2011; 5:199-208. [PMID: 21141979 DOI: 10.1021/nn1023522] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
Amphotericin B (AMB) has long been considered the most effective drug in the treatment of serious invasive fungal infections. There are, however, major limitations to its use, due to several adverse effects, including acute infusional reactions and, most relevant, a dose-dependent nephrotoxicity. At least some of these effects are attributed to the aggregation of AMB as a result of its poor water solubility. To overcome this problem, reformulated versions of the drug have been developed, including a micellar dispersion of AMB with sodium deoxycholate (AMBD), its encapsulation into liposomes, or its incorporation into lipidic complexes. The development of nanobiotechnologies provides novel potential drug delivery systems that make use of nanomaterials such as functionalized carbon nanotubes (f-CNTs), which are emerging as an innovative and efficient tool for the transport and cellular translocation of therapeutic molecules. In this study, we prepared two conjugates between f-CNTs and AMB. The antifungal activity of these conjugates was tested against a collection of reference and clinical fungal strains, in comparison to that of AMB alone or AMBD. Measured minimum inhibition concentration (MIC) values for f-CNT-AMB conjugates were either comparable to or better than those displayed by AMB and AMBD. Furthermore, AMBD-resistant Candida strains were found to be susceptible to f-CNT-AMB 1. Additional studies, aimed at understanding the mechanism of action of the conjugates, suggest a nonlytic mechanism, since the compounds show a major permeabilizing effect on the tested fungal strains only after extended incubation. Interestingly, the f-CNT-AMB 1 does not show any significant toxic effect on Jurkat cells at antifungal concentrations.
Collapse
|
17
|
Abstract
Echinocandins are the most recent introduction to the antifungal armamentarium and target the synthesis of β-(1,3)-glucan, the major structural polysaccharide of the fungal cell wall. Mechanisms have been identified that reduce the efficacy of the echinocandins: mutations of the Fks subunit of the target enzyme complex or a compensatory increase in the production of chitin, the second structural cell wall polysaccharide.
Collapse
Affiliation(s)
- Carol A Munro
- Aberdeen Fungal Group, School of Medical Sciences, Institute of Medical Sciences, University of AberdeenForesterhill, Aberdeen, AB25 2ZDUK
| |
Collapse
|
18
|
Pal S, Yoon EJ, Park SH, Choi EC, Song JM. Metallopharmaceuticals based on silver(I) and silver(II) polydiguanide complexes: activity against burn wound pathogens. J Antimicrob Chemother 2010; 65:2134-40. [DOI: 10.1093/jac/dkq294] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
19
|
In vitro fungicidal activities of echinocandins against Candida metapsilosis, C. orthopsilosis, and C. parapsilosis evaluated by time-kill studies. Antimicrob Agents Chemother 2010; 54:2194-7. [PMID: 20145083 DOI: 10.1128/aac.01538-09] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Anidulafungin, micafungin, and caspofungin in vitro activities against Candida metapsilosis, C. orthopsilosis, and C. parapsilosis were evaluated by MICs and time-kill methods. All echinocandins showed lower MICs (mean MICs, 0.05 to 0.71 mg/liter) and the highest killing rates (-0.06 to -0.05 CFU/ml/h) for C. metapsilosis and C. orthopsilosis rather than for C. parapsilosis (mean MICs, 0.59 to 1.68 mg/liter). Micafungin and anidulafungin killing rates were greater than those determined for caspofungin. None of the echinocandins had fungicidal activity against C. parapsilosis.
Collapse
|
20
|
Walker LA, Gow NA, Munro CA. Fungal echinocandin resistance. Fungal Genet Biol 2010; 47:117-26. [PMID: 19770064 PMCID: PMC2812698 DOI: 10.1016/j.fgb.2009.09.003] [Citation(s) in RCA: 197] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2009] [Revised: 08/19/2009] [Accepted: 09/09/2009] [Indexed: 11/21/2022]
Abstract
The echinocandins are the newest class of antifungal agents in the clinical armory. These secondary metabolites are non-competitive inhibitors of the synthesis of beta-(1,3)-glucan, a major structural component of the fungal cell wall. Recent work has shown that spontaneous mutations can arise in two hot spot regions of Fks1 the target protein of echinocandins that reduce the enzyme's sensitivity to the drug. However, other strains have been isolated in which the sequence of FKS1 is unaltered yet the fungus has decreased sensitivity to echinocandins. In addition it has been shown that echinocandin-treatment can induce cell wall salvage mechanisms that result in the compensatory upregulation of chitin synthesis in the cell wall. This salvage mechanism strengthens cell walls damaged by exposure to echinocandins. Therefore, fungal resistance to echinocandins can arise due to the selection of either stable mutational or reversible physiological alterations that decrease susceptibility to these antifungal agents.
Collapse
Affiliation(s)
| | | | - Carol A. Munro
- School of Medical Sciences, University of Aberdeen, Aberdeen, AB25 2ZD, UK
| |
Collapse
|
21
|
Nguyen KT, Ta P, Hoang BT, Cheng S, Hao B, Nguyen MH, Clancy CJ. Characterising the post-antifungal effects of micafungin against Candida albicans, Candida glabrata, Candida parapsilosis and Candida krusei isolates. Int J Antimicrob Agents 2010; 35:80-4. [DOI: 10.1016/j.ijantimicag.2009.09.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2009] [Revised: 08/31/2009] [Accepted: 09/02/2009] [Indexed: 10/20/2022]
|
22
|
Lockwood AM, Wyatt KN, Abraham P, Montgomery SP, Macleod J. Candida Guilliermondii Fungemia in a Critically III Trauma Patient. J Pharm Technol 2010. [DOI: 10.1177/875512251002600106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Objective: To report a case of fungemia caused by Candida guilliermondii in a non-neutropenic 17-year-old critically ill trauma patient. Case Summary: Few case reports and small surveys have reported invasive infection caused by C. guilliermondii, most of which has occurred in immunocompromised populations. We present the case of a 17-year-old trauma patient who developed fungemia with C. guilliermondii during his intensive care unit stay. While he had multiple risk factors for the development of candidemia, this was an atypical finding, considering that this patient was not neutropenic. Due to the variable resistance patterns with this species of Candida, mean inhibitory concentration testing was requested, which demonstrated susceptibility to fluconazole. The patient was thus treated with fluconazole and his fungemia resolved without relapse. Discussion: C. guilliermondii accounts for 1–5% of cases of non- albicans Candida. While C. guilliermondii typically does not cause invasive infection, it has been associated with severe, life-threatening candidiasis in immunocompromised patients. Due to its low incidence, guidelines for the management of C. guilliermondii infections have not been well established and specific risk factors have not been identified. While risk factors specific to C. guilliermondii may mimic established risk factors for candidal fungemia in general, an important aspect to consider in the treatment of this fungus is its unique resistance profile compared with other non- albicans species. Conclusions: This case illustrates the development of an uncommon fungal pathogen in a non-immunocompromised host, compared with previously published cases series. Susceptibility testing is recommended to prevent therapeutic failures due to varying resistance patterns with C. guilliermondii.
Collapse
Affiliation(s)
- Alissa M Lockwood
- ALISSA M LOCKWOOD PharmD, Clinical Pharmacist Specialist, Critical Care, Department of Pharmacy and Drug Information, Parkland Health & Hospital System, Dallas, TX
| | - Katleen N Wyatt
- KATLEEN N WYATT PharmD BCPS, Clinical Pharmacist Specialist, Critical Care, Department of Pharmacy and Drug Information, Grady Health System, Atlanta, GA
| | - Prasad Abraham
- PRASAD ABRAHAM PharmD BCPS, Clinical Pharmacist Specialist, Critical Care, Department of Pharmacy and Drug Information, Grady Health System
| | - Sean P Montgomery
- MAJORSEAN P MONT-GOMERYMD MC FACS, Trauma and Critical Care Surgeon, General Surgery Service, Department of Surgery, Walter Reed Army Medical Center, Washington, DC
| | - Jana Macleod
- JANA MACLEOD MD FACS, Assistant Professor of Surgery, Department of Surgery, Emory University School of Medicine
| |
Collapse
|
23
|
Comparison of anidulafungin MICs determined by the clinical and laboratory standards institute broth microdilution method (M27-A3 document) and Etest for Candida species isolates. Antimicrob Agents Chemother 2009; 54:1347-50. [PMID: 20028814 DOI: 10.1128/aac.01324-09] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Anidulafungin Etest and CLSI MICs were compared for 143 Candida sp. isolates to assess essential (within 2 log(2) dilutions) and categorical agreements (according to three susceptibility breakpoints). Based on agreement percentages, our data indicated that Etest is not suitable to test anidulafungin against Candida parapsilosis and C. guilliermondii (54.4 to 82.4% essential and categorical agreements) but is more suitable for C. albicans, C. glabrata, C. krusei, and C. tropicalis (87.9 to 100% categorical agreement).
Collapse
|
24
|
Carter NJ, Keating GM. Micafungin: a review of its use in the prophylaxis and treatment of invasive Candida infections in pediatric patients. Paediatr Drugs 2009; 11:271-91. [PMID: 19566111 DOI: 10.2165/00148581-200911040-00006] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Intravenous micafungin (Mycamine; Funguard) is an echinocandin indicated in Japan and the EU for the treatment of pediatric patients (including neonates) with invasive candidiasis and as prophylaxis against Candida infection in pediatric patients undergoing allogeneic hematopoietic stem cell transplantation (HSCT). In the EU, micafungin is also indicated in pediatric patients who are expected to have neutropenia for >/=10 days. In Japan, children may also receive micafungin for the treatment of, or as prophylaxis against, invasive Aspergillus infection. Micafungin is not currently approved for use in pediatric patients in the US. Micafungin has very good antifungal activity against a wide range of Candida spp. in vitro. It has a favorable pharmacokinetic profile allowing for once-daily administration, has few drug-drug interactions, and reports of resistance are rare. The results of pediatric substudies indicate that intravenous micafungin is effective in a majority of patients for the treatment of candidemia and other types of invasive candidiasis, and provides effective prophylaxis against invasive fungal infections in pediatric patients undergoing HSCT. The tolerability profile of micafungin in pediatric patients was generally acceptable. In the EU, micafungin is indicated for use when other antifungal medications are not appropriate. Therefore, micafungin provides an alternative to other antifungal agents used in the management of candidemia and invasive candidiasis in pediatric patients, or as prophylaxis against fungal infections in pediatric patients undergoing HSCT.
Collapse
Affiliation(s)
- Natalie J Carter
- Wolters Kluwer Health mid R: Adis, Auckland, New Zealand, an editorial office of Wolters Kluwer Health, Philadelphia, Pennsylvania, USA.
| | | |
Collapse
|
25
|
Pemán J, Cantón E, Espinel-Ingroff A. Antifungal drug resistance mechanisms. Expert Rev Anti Infect Ther 2009; 7:453-60. [PMID: 19400764 DOI: 10.1586/eri.09.18] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Antifungal resistance is a prominent feature in the management of invasive mycoses, with important implications for morbidity and mortality. Microbiological resistance, the most common cause of refractory infection, is associated with a fungal pathogen for which an antifungal MIC is higher than average or within the range designated as the resistant breakpoint. Four major mechanisms of resistance to azoles have been described in Candida spp.: decreased intracellular drug concentration by activation of efflux systems or reduction of drug penetration, modification of the target site, upregulation of the target enzyme and development of bypass pathways. Conversely, echinocandins are a poor substrate for multidrug efflux transporters, and their mechanisms of resistance are associated with point mutations and/or overexpression of FKS1 and FKS2 genes. Acquired resistance to flucytosine results from defects in its metabolism through enzymatic mutations, whereas resistance to amphotericin B may be mediated by increased catalase activity or defects in ergosterol biosynthesis.
Collapse
Affiliation(s)
- Javier Pemán
- Servicio de Microbiología, Hospital Universitario La Fe, Avenida Campanar 21, 46009 Valencia, Spain.
| | | | | |
Collapse
|
26
|
Quindós G, Eraso E, Javier Carrillo-Muñoz A, Cantón E, Pemán J. Actividad antifúngica in vitro de la micafungina. Rev Iberoam Micol 2009; 26:35-41. [DOI: 10.1016/s1130-1406(09)70006-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2009] [Accepted: 02/09/2009] [Indexed: 11/25/2022] Open
|
27
|
de Aquino Lemos J, Costa CR, de Araújo CR, Souza LKHE, Silva MDRR. Susceptibility testing of Candida albicans isolated from oropharyngeal mucosa of HIV(+) patients to fluconazole, amphotericin B and Caspofungin. killing kinetics of caspofungin and amphotericin B against fluconazole resistant and susceptible isolates. Braz J Microbiol 2009; 40:163-9. [PMID: 24031337 PMCID: PMC3768489 DOI: 10.1590/s1517-838220090001000028] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2007] [Revised: 03/09/2008] [Accepted: 02/17/2009] [Indexed: 11/22/2022] Open
Abstract
A clear understanding of the pharmacodynamic properties of antifungal agents is important for the adequate treatment of fungal infections like candidiasis. For certain antifungal agents, the determination of Minimal Fungicidal Concentration (MFC) and time kill curve could be clinically more relevant than the determination of the Minimal Inhibitory Concentration (MIC). In this study, MIC and MFC to fluconazole, amphotericin B and caspofungin against C. albicans isolates and the killing patterns obtained with caspofungin and amphotericin B against susceptible and resistant strains to fluconazole were determined. The results of MICs showed that all C. albicans isolates were highly susceptible to amphotericin B, but two isolates were fluconazole resistant. The comparative analysis between MIC and MFC showed that MFC of fluconazole was fourfold higher than MIC in 41.9% of the C. albicans isolates. Same values of MFC and MIC of amphotericin B and caspofungin were found for 71% of the isolates. Correlation between time kill curves and MFC of amphotericin B and caspofungin against all 4 isolates tested was observed. The caspofungin killing effect was more evident at MFC in 6 hours of incubation than at MIC in this time, suggesting dependence of concentration. The similarity of results of time-kill curve and MFC values indicate that determination of MFC is an alternative for the detection of the fungicidal activity of these drugs.
Collapse
Affiliation(s)
- Janine de Aquino Lemos
- Instituto de Patologia Tropical e Saúde Pública da Universidade Federal de Goiás , Goiínia, GO , Brasil
| | | | | | | | | |
Collapse
|
28
|
Abstract
Antifungal susceptibility testing is a very dynamic field of medical mycology. Standardization of in vitro susceptibility tests by the Clinical and Laboratory Standards Institute (CLSI) and the European Committee for Antimicrobial Susceptibility Testing (EUCAST), and current availability of reference methods constituted the major remarkable steps in the field. Based on the established minimum inhibitory concentration (MIC) breakpoints, it is now possible to determine the susceptibilities of Candida strains to fluconazole, itraconazole, voriconazole, and flucytosine. Moreover, utility of fluconazole antifungal susceptibility tests as an adjunct in optimizing treatment of candidiasis has now been validated. While the MIC breakpoints and clinical significance of susceptibility testing for the remaining fungi and antifungal drugs remain yet unclear, modifications of the available methods as well as other methodologies are being intensively studied to overcome the present drawbacks and limitations. Among the other methods under investigation are Etest, colorimetric microdilution, agar dilution, determination of fungicidal activity, flow cytometry, and ergosterol quantitation. Etest offers the advantage of practical application and favorable agreement rates with the reference methods that are frequently above acceptable limits. However, MIC breakpoints for Etest remain to be evaluated and established. Development of commercially available, standardized colorimetric panels that are based on CLSI method parameters has added more to the antifungal susceptibility testing armamentarium. Flow cytometry, on the other hand, appears to offer rapid susceptibility testing but requires specified equipment and further evaluation for reproducibility and standardization. Ergosterol quantitation is another novel approach, which appears potentially beneficial particularly in discrimination of azole-resistant isolates from heavy trailers. The method is yet investigational and requires to be further studied. Developments in methodology and applications of antifungal susceptibility testing will hopefully provide enhanced utility in clinical guidance of antifungal therapy. However, and particularly in immunosuppressed host, in vitro susceptibility is and will remain only one of several factors that influence clinical outcome.
Collapse
Affiliation(s)
- Sevtap Arikan
- Department of Microbiology and Clinical Microbiology, Hacettepe University Medical School, Ankara, Turkey.
| |
Collapse
|
29
|
Killing of Neisseria gonorrhoeae, Streptococcus agalactiae (group B streptococcus), Haemophilus ducreyi, and vaginal Lactobacillus by 3-O-octyl-sn-glycerol. Antimicrob Agents Chemother 2008; 52:1577-9. [PMID: 18227178 DOI: 10.1128/aac.01023-07] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The microbicide candidate octylglycerol inactivates sexually transmitted bacterial pathogens at concentrations which spare normal vaginal flora (lactobacillus). Standard minimum microbicidal concentration assays and time-kill assays revealed the drug concentrations and times required for inactivation. Octylglycerol concentrations must exceed the binding capacity of any human serum albumin to be effective.
Collapse
|
30
|
Impact of pharmacodynamics and pharmacokinetics on echinocandin dosing strategies. Curr Opin Infect Dis 2007; 20:587-91. [DOI: 10.1097/qco.0b013e3282f1bea3] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
31
|
Pemán J, Bosch M, Cantón E, Viudes A, Jarque I, Gómez-García M, García-Martínez JM, Gobernado M. Fungemia due to Candida guilliermondii in a pediatric and adult population during a 12-year period. Diagn Microbiol Infect Dis 2007; 60:109-12. [PMID: 17889486 DOI: 10.1016/j.diagmicrobio.2007.07.014] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2007] [Revised: 07/11/2007] [Accepted: 07/21/2007] [Indexed: 11/28/2022]
Abstract
Candida guilliermondii fungemia is usually described in adults with hematologic malignancies, but in children, only 2 episodes have been published. From 1995 to 2006, 7 episodes (5 in children) were detected in our hospital. Molecular typing excluded a common infection source. C. guilliermondii fungemia may occur in children with underlying conditions other than cancer.
Collapse
Affiliation(s)
- Javier Pemán
- Servicio de Microbiología, Hospital Universitario la Fe, Valencia 46009, Spain.
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Aperis G, Myriounis N, Spanakis EK, Mylonakis E. Developments in the treatment of candidiasis: more choices and new challenges. Expert Opin Investig Drugs 2006; 15:1319-36. [PMID: 17040194 DOI: 10.1517/13543784.15.11.1319] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
The incidence of oesophageal candidiasis, candidaemia and disseminated candidiasis has increased dramatically. In addition to the amphotericin B formulations and fluconazole, the echinocandins anidulafungin, caspofungin and micafungin and the newer triazoles posaconazole and voriconazole are in the last stages of development and are becoming available for the management of candidiasis. This review presents these new agents and addresses their role in the treatment of candidiasis. All new antifungal agents exhibit potent activity against Candida spp. and echinocandins are fungicidal against most Candida spp. but appear to be less potent against certain species, such as Candida parapsilosis and C. guilliermondii. Systemic antifungal therapy can now be individualised based on the severity of the infection, comorbid conditions and the Candida spp. causing the infection. Studies are needed to investigate the possible development of resistance and the efficacy of these antifungal agents against the more resistant Candida spp.
Collapse
Affiliation(s)
- George Aperis
- Massachusetts General Hospital, Division of Infectious Diseases, Gray-Jackson 504, 55 Fruit Street, Boston, MA 02114, USA
| | | | | | | |
Collapse
|