1
|
Reddy CA, McGowan E, Yadlapati R, Peterson K. AGA Clinical Practice Update on Esophageal Dysfunction Due to Disordered Immunity and Infection: Expert Review. Clin Gastroenterol Hepatol 2024:S1542-3565(24)00826-7. [PMID: 39436337 DOI: 10.1016/j.cgh.2024.08.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Revised: 07/18/2024] [Accepted: 08/09/2024] [Indexed: 10/23/2024]
Abstract
METHODS This expert review was commissioned and approved by the American Gastroenterological Association (AGA) Institute Clinical Practice Updates Committee (CPUC) and the AGA Governing Board to provide timely guidance on a topic of high clinical importance to the AGA membership, and underwent internal peer review by the CPUC and external peer review through standard procedures of Clinical Gastroenterology and Hepatology. These Best Practice Advice (BPA) statements were drawn from a review of the published literature and from expert opinion. Since systematic reviews were not performed, these BPA statements do not carry formal ratings regarding the quality of evidence or strength of the presented considerations. DESCRIPTION Infectious and immune-mediated esophageal disorders are poorly understood and often under-diagnosed conditions that lead to esophageal dysfunction and health care costs due to repeated procedures and a lack of understanding of their etiology and pathogenesis. Without a high index of suspicion, these disorders may be overlooked. Esophageal dysfunction may arise from active, localized infection and immune-mediated disease (ie, candida, etc.) or from an organ-specific manifestation of a more diffuse immune-mediated disease or infection (ie, systemic sclerosis, connective tissue disease, neurologic disease). These conditions can sometimes lead to neuromuscular dysfunction and subsequent esophageal dysmotility. Awareness of local and systemic processes that lead to esophageal dysfunction will improve patient outcomes by focusing therapeutics and limiting unnecessary procedures. Therefore, the purpose of this AGA Clinical Practice Update Expert Review is to provide BPA on diagnostic considerations of immune-mediated disorders that should be considered when encountering patients with dysphagia, heartburn, and odynophagia. Best Practice Advice Statements: BEST PRACTICE ADVICE 1: Gastroenterologists should be aware of the esophageal manifestations of systemic immunologic and infectious diseases to reduce diagnostic delay. Clinicians should identify if there are risks for inflammatory or infectious possibilities for a patient's esophageal symptoms and investigate for these disorders as a potential cause of esophageal dysfunction. BEST PRACTICE ADVICE 2: Once esophageal infection is identified, clinicians should identify whether accompanying signs/symptoms suggest immunocompromise leading to a more systemic infection. Consultation with an infectious disease expert will aid in guiding appropriate treatment. BEST PRACTICE ADVICE 3: If symptoms do not improve after therapy for infectious esophagitis, evaluation for refractory infection or additional underlying sources of esophageal and immunologic dysfunction should be performed. BEST PRACTICE ADVICE 4: In individuals with eosinophilic esophagitis (EoE) who continue to experience symptoms of esophageal dysfunction despite histologic and endoscopic disease remission, clinicians should be aware that some patients with EoE may develop motility disorders. Further evaluation of esophageal motility may be warranted. BEST PRACTICE ADVICE 5: In individuals with histologic and endoscopic features of lymphocytic esophagitis, clinicians should consider treatment of lymphocytic-related inflammation with proton-pump inhibitor therapy or swallowed topical corticosteroids and as needed esophageal dilation. BEST PRACTICE ADVICE 6: In patients who present with esophageal symptoms in the setting of hypereosinophilia (absolute eosinophil count [AEC] >1500 cells/uL), consider further work-up of non-EoE eosinophilic gastrointestinal (GI) disease, hypereosinophilic syndrome, and eosinophilic granulomatosis with polyangiitis (EGPA). Consultation with allergy/immunology may help guide further diagnostic work-up and treatment. BEST PRACTICE ADVICE 7: In individuals with rheumatologic diseases of systemic sclerosis (SSc), mixed connective tissue disease (MCTD), systemic lupus erythematosus (SLE), or Sjogren's disease, clinicians should be aware that esophageal symptoms can occur due to involvement of the esophageal muscle layer, resulting in dysmotility and/or incompetence of the lower esophageal sphincter. The degree of dysfunction is often especially significant in those with SSc or MCTD. BEST PRACTICE ADVICE 8: In individuals with Crohn's disease, clinicians should be aware that a minority of individuals can develop esophageal involvement from inflammatory, stricturing, or fistulizing changes with granulomas seen histologically. Esophageal manifestations of Crohn's disease tend to occur in individuals with active intestinal disease. BEST PRACTICE ADVICE 9: In individuals with dermatologic diseases of lichen planus or bullous disorders, clinicians should be aware that dysphagia can occur due to endoscopically visible esophageal mucosal involvement. Esophageal lichen planus, in particular, can occur without skin involvement and can be difficult to define on esophageal histopathology. BEST PRACTICE ADVICE 10: Clinicians should consider infectious and inflammatory causes of secondary achalasia during initial evaluation. One should query for any history of recent COVID infections, risks for Chagas disease, and symptoms or signs of eosinophilic disease.
Collapse
Affiliation(s)
- Chanakyaram A Reddy
- Center for Esophageal Diseases, Division of Gastroenterology, Baylor University Medical Center, Dallas, Texas
| | - Emily McGowan
- Division of Allergy and Immunology, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Rena Yadlapati
- Division of Gastroenterology, UCSD Center for Esophageal Diseases, GI Motility Lab, University of California San Diego, GEODE Research Program, San Francisco, California
| | - Kathryn Peterson
- Division of Gastroenterology, University of Utah, Salt Lake City, Utah.
| |
Collapse
|
2
|
Ortiz B, Varela D, Fontecha G, Torres K, Cornely OA, Salmanton-García J. Strengthening Fungal Infection Diagnosis and Treatment: An In-depth Analysis of Capabilities in Honduras. Open Forum Infect Dis 2024; 11:ofae578. [PMID: 39421702 PMCID: PMC11483579 DOI: 10.1093/ofid/ofae578] [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: 07/11/2024] [Accepted: 09/30/2024] [Indexed: 10/19/2024] Open
Abstract
Background Invasive fungal infections (IFIs) are a major public health concern in low- and middle-income countries (LMICs) due to limited diagnostic and treatment resources, leading to high morbidity and mortality. Despite their significant global burden, IFIs are underrecognized and underdiagnosed in LMICs. This study evaluates the diagnostic and therapeutic capacities for managing IFI in Honduras, a country with unique health care challenges. Methods From March to December 2023, a comprehensive survey was conducted across multiple health care centers in Honduras. The survey, reviewed for content and clarity by local medical institutions, targeted medical microbiologists and clinicians to assess various aspects of fungal disease diagnosis and treatment. Data included the availability and use of diagnostic tools and antifungal therapies, identifying gaps and limitations in current practices. Results The survey revealed that Candida spp (97.4%) and Aspergillus spp (35.9%) were the most concerning pathogens. Although microscopy and culture methods were available in most institutions, their application in suspected IFI cases was inconsistent, and antifungal susceptibility testing was rarely performed. Advanced diagnostic techniques, such as antigen detection, were available in only a few institutions, while antibody detection and polymerase chain reaction testing were entirely absent. All hospitals had access to at least 1 triazole antifungal, typically fluconazole, but there was a notable scarcity of more potent antifungals, including amphotericin B formulations and echinocandins. The limited use of available diagnostic tools and the restricted availability of essential antifungals were identified as major barriers to effective IFI management. Conclusions This study highlights significant gaps in the diagnostic and therapeutic capabilities for managing IFI in Honduras. The underutilization of basic diagnostic tools, the inaccessibility of advanced testing methods, and the limited availability of essential antifungal medications underscore the urgent need for capacity-building initiatives, infrastructure improvements, and policy reforms. Addressing these deficiencies is critical for enhancing the management of IFI in Honduras, with broader implications for similar LMIC settings. These findings can inform targeted interventions and resource allocation to improve outcomes for patients with IFI.
Collapse
Affiliation(s)
- Bryan Ortiz
- Instituto de Investigaciones en Microbiología, Facultad de Ciencias, Universidad Nacional Autónoma de Honduras, Tegucigalpa, Honduras
| | - Diana Varela
- Servicio de Infectología, Servicio de Atención Integral de Pacientes con VIH, Hospital Escuela, Tegucigalpa, Honduras
- Instituto de Enfermedades Infecciosas y Parasitarias Antonio Vidal, Tegucigalpa, Honduras
| | - Gustavo Fontecha
- Instituto de Investigaciones en Microbiología, Facultad de Ciencias, Universidad Nacional Autónoma de Honduras, Tegucigalpa, Honduras
| | - Karla Torres
- Agrupación de Microbiólogos Propietarios de Laboratorios Privados de Honduras, Tegucigalpa, Honduras
- Departamento de Química y Biología, Centro Universitario Regional de Occidente, Santa Rosa de Copán, Honduras
| | - Oliver A Cornely
- Faculty of Medicine and University Hospital Cologne, Institute of Translational Research, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, University of Cologne, Cologne, Germany
- Faculty of Medicine and University Hospital Cologne, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf and Excellence Center for Medical Mycology, University of Cologne, Cologne, Germany
- German Centre for Infection Research, Partner Site Bonn-Cologne, Cologne, Germany
- Faculty of Medicine and University Hospital Cologne, Clinical Trials Centre Cologne, University of Cologne, Cologne, Germany
| | - Jon Salmanton-García
- Faculty of Medicine and University Hospital Cologne, Institute of Translational Research, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, University of Cologne, Cologne, Germany
- Faculty of Medicine and University Hospital Cologne, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf and Excellence Center for Medical Mycology, University of Cologne, Cologne, Germany
- German Centre for Infection Research, Partner Site Bonn-Cologne, Cologne, Germany
| |
Collapse
|
3
|
Tseng KY, Huang YT, Huang YT, Su YT, Wang AN, Weng WY, Ke CL, Yeh YC, Wang JJ, Du SH, Gu ZQ, Chen WL, Lin CH, Tsai YH. Regulation of candidalysin underlies Candida albicans persistence in intravascular catheters by modulating NETosis. PLoS Pathog 2024; 20:e1012319. [PMID: 38885290 PMCID: PMC11213320 DOI: 10.1371/journal.ppat.1012319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 06/28/2024] [Accepted: 06/06/2024] [Indexed: 06/20/2024] Open
Abstract
Candida albicans is a leading cause of intravascular catheter-related infections. The capacity for biofilm formation has been proposed to contribute to the persistence of this fungal pathogen on catheter surfaces. While efforts have been devoted to identifying microbial factors that modulate C. albicans biofilm formation in vitro, our understanding of the host factors that may shape C. albicans persistence in intravascular catheters is lacking. Here, we used multiphoton microscopy to characterize biofilms in intravascular catheters removed from candidiasis patients. We demonstrated that, NETosis, a type of neutrophil cell death with antimicrobial activity, was implicated in the interaction of immune cells with C. albicans in the catheters. The catheter isolates exhibited reduced filamentation and candidalysin gene expression, specifically in the total parenteral nutrition culture environment. Furthermore, we showed that the ablation of candidalysin expression in C. albicans reduced NETosis and conferred resistance to neutrophil-mediated fungal biofilm elimination. Our findings illustrate the role of neutrophil NETosis in modulating C. albicans biofilm persistence in an intravascular catheter, highlighting that C. albicans can benefit from reduced virulence expression to promote its persistence in an intravascular catheter.
Collapse
Affiliation(s)
- Kuo-Yao Tseng
- Laboratory of Host–Microbe Interactions and Cell Dynamics, Institute of Microbiology and Immunology, National Yang Ming Chiao Tung University, Taipei, 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
- Graduate Institute of Clinical Laboratory Sciences and Medical Biotechnology, National Taiwan University, Taipei, Taiwan
| | - Yu-Ting Huang
- Laboratory of Host–Microbe Interactions and Cell Dynamics, Institute of Microbiology and Immunology, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yu-Ting Su
- Laboratory of Host–Microbe Interactions and Cell Dynamics, Institute of Microbiology and Immunology, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - An-Ni Wang
- Laboratory of Host–Microbe Interactions and Cell Dynamics, Institute of Microbiology and Immunology, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Wen-Yen Weng
- Laboratory of Host–Microbe Interactions and Cell Dynamics, Institute of Microbiology and Immunology, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Cai-Ling Ke
- Department of Biochemical Science and Technology, College of Life Science, National Taiwan University, Taipei, Taiwan
| | - Yu-Chiao Yeh
- Department of Biochemical Science and Technology, College of Life Science, National Taiwan University, Taipei, Taiwan
| | - Jhih-Jie Wang
- Laboratory of Host–Microbe Interactions and Cell Dynamics, Institute of Microbiology and Immunology, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Shin-Hei Du
- Department of Laboratory Medicine, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Zi-Qi Gu
- Laboratory of Host–Microbe Interactions and Cell Dynamics, Institute of Microbiology and Immunology, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Wei-Lin Chen
- Laboratory of Host–Microbe Interactions and Cell Dynamics, Institute of Microbiology and Immunology, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Program in Molecular Medicine, National Yang Ming Chiao Tung University and Academia Sinica, Taipei, Taiwan
| | - Ching-Hsuan Lin
- Department of Biochemical Science and Technology, College of Life Science, National Taiwan University, Taipei, Taiwan
| | - Yu-Huan Tsai
- Laboratory of Host–Microbe Interactions and Cell Dynamics, Institute of Microbiology and Immunology, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Program in Molecular Medicine, National Yang Ming Chiao Tung University and Academia Sinica, Taipei, Taiwan
- Center for Molecular and Clinical Immunology, Chang Gung University, Taoyuan, Taiwan
| |
Collapse
|
4
|
Keyvanfar A, Najafiarab H, Talebian N, Tafti MF, Adeli G, Ghasemi Z, Tehrani S. Drug-resistant oral candidiasis in patients with HIV infection: a systematic review and meta-analysis. BMC Infect Dis 2024; 24:546. [PMID: 38822256 PMCID: PMC11143751 DOI: 10.1186/s12879-024-09442-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 05/27/2024] [Indexed: 06/02/2024] Open
Abstract
BACKGROUND Oral candidiasis (OC) is a prevalent opportunistic infection in patients with human immunodeficiency virus (HIV) infection. The increasing resistance to antifungal agents in HIV-positive individuals suffering from OC raised concerns. Thus, this study aimed to investigate the prevalence of drug-resistant OC in HIV-positive patients. METHODS Pubmed, Web of Science, Scopus, and Embase databases were systematically searched for eligible articles up to November 30, 2023. Studies reporting resistance to antifungal agents in Candida species isolated from HIV-positive patients with OC were included. Baseline characteristics, clinical features, isolated Candida species, and antifungal resistance were independently extracted by two reviewers. The pooled prevalence with a 95% confidence interval (CI) was calculated using the random effect model or fixed effect model. RESULTS Out of the 1942 records, 25 studies consisting of 2564 Candida species entered the meta-analysis. The pooled prevalence of resistance to the antifungal agents was as follows: ketoconazole (25.5%, 95% CI: 15.1-35.8%), fluconazole (24.8%, 95% CI: 17.4-32.1%), 5-Flucytosine (22.9%, 95% CI: -13.7-59.6%), itraconazole (20.0%, 95% CI: 10.0-26.0%), voriconazole (20.0%, 95% CI: 1.9-38.0%), miconazole (15.0%, 95% CI: 5.1-26.0%), clotrimazole (13.4%, 95% CI: 2.3-24.5%), nystatin (4.9%, 95% CI: -0.05-10.3%), amphotericin B (2.9%, 95% CI: 0.5-5.3%), and caspofungin (0.1%, 95% CI: -0.3-0.6%). Furthermore, there were high heterogeneities among almost all included studies regarding the resistance to different antifungal agents (I2 > 50.00%, P < 0.01), except for caspofungin (I2 = 0.00%, P = 0.65). CONCLUSIONS Our research revealed that a significant number of Candida species found in HIV-positive patients with OC were resistant to azoles and 5-fluocytosine. However, most of the isolates were susceptible to nystatin, amphotericin B, and caspofungin. This suggests that initial treatments for OC, such as azoles, may not be effective. In such cases, healthcare providers may need to consider prescribing alternative treatments like polyenes and caspofungin. REGISTRATION The study protocol was registered in the International Prospective Register of Systematic Reviews as PROSPERO (Number: CRD42024497963).
Collapse
Affiliation(s)
- Amirreza Keyvanfar
- Infectious Diseases and Tropical Medicine Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hanieh Najafiarab
- Preventative Gynecology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Niki Talebian
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahdi Falah Tafti
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Gelareh Adeli
- Faculty of Biological Sciences, North Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Zahra Ghasemi
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Shabnam Tehrani
- Infectious Diseases and Tropical Medicine Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
5
|
Salmanton-García J, Hoenigl M, Salzer HJF, Lackner M, Prattes J, Dichtl K, Winkler-Zamani M, Krause R, Stemler J, Lass-Flörl C, Cornely OA, Willinger B. The Austrian landscape of diagnostic capacity and access to treatment for invasive fungal infections. Mycoses 2023; 66:1056-1063. [PMID: 37592370 DOI: 10.1111/myc.13650] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Revised: 07/31/2023] [Accepted: 08/04/2023] [Indexed: 08/19/2023]
Abstract
INTRODUCTION Immunosuppression after chemotherapy, stem cell transplantation or solid organ transplantation are the main risk factors for invasive fungal infections in Austria. Here, we aim to describe the status of laboratory mycology and the access to antifungal treatment in Austria. METHODS Between October and November 2021, hospitals were contacted to participate in our online survey: www.clinicalsurveys.net/uc/IFI_management_capacity/. Centres were required to provide information on their institutional profile; self-assessment of burden of invasive fungal infections; access to microscopy, culture, serology, antigen detection and molecular testing; and availability of antifungal agents and therapeutic drug monitoring. RESULTS Responses were collected from university hospitals and laboratories in Graz, Innsbruck, Linz and Vienna. The four hospitals can provide tertiary care and were highly specialised, including management of patients with severe immunosuppression. All sites consider the incidence of invasive fungal infections to be moderate. Access to microscopy, culture, serology, antigen detection and molecular testing is provided regardless of laboratory. The maximum capacity to identify fungi varies from institution to institution. All currently marketed antifungal agents are available at the four sites. CONCLUSION Austria is currently well equipped to deal with the emerging threat of invasive fungal infections. However, hospitals may consider preparing for the potential endemicity of certain infections in the near future.
Collapse
Affiliation(s)
- Jon Salmanton-García
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Faculty of Medicine and University Hospital 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
- German Centre for Infection Research (DZIF), Partner Site Bonn-Cologne, Cologne, Germany
| | - Martin Hoenigl
- Department of Internal Medicine, Division of Infectious Diseases, Medical University of Graz, Austria
- BioTechMed, Austria
| | - Helmut J F Salzer
- Department of Internal Medicine 4 - Pneumology, Kepler University Hospital, Linz, Austria
- Medical Faculty, Johannes Kepler University Linz, Linz, Austria
| | - Michaela Lackner
- Institute of Hygiene and Medical Microbiology, Medical University of Innsbruck, European Confederation of Medical Mycology Excellence Centre in Fungal Infections, Innsbruck, Austria
| | - Juergen Prattes
- Department of Internal Medicine, Division of Infectious Diseases, Medical University of Graz, Austria
- BioTechMed, Austria
| | - Karl Dichtl
- Diagnostic and Research Institute of Hygiene, Microbiology and Environmental Medicine, Medical University of Graz, Austria
| | - Markus Winkler-Zamani
- Institute of Clinical Pathology and Molecular Pathology, Kepler University Hospital and Johannes Kepler University, Linz, Austria
| | - Robert Krause
- Department of Internal Medicine, Division of Infectious Diseases, Medical University of Graz, Austria
- BioTechMed, Austria
| | - Jannik Stemler
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Faculty of Medicine and University Hospital 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
- German Centre for Infection Research (DZIF), Partner Site Bonn-Cologne, Cologne, Germany
| | - Cornelia Lass-Flörl
- Institute of Hygiene and Medical Microbiology, Medical University of Innsbruck, European Confederation of Medical Mycology Excellence Centre in Fungal Infections, Innsbruck, Austria
| | - Oliver A Cornely
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Faculty of Medicine and University Hospital 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
- German Centre for Infection Research (DZIF), Partner Site Bonn-Cologne, Cologne, Germany
- Clinical Trials Centre Cologne (ZKS Köln), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Birgit Willinger
- Department of Laboratory Medicine, Division of Clinical Microbiology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
6
|
Sharma B, Nonzom S. Mucormycosis and Its Upsurge During COVID-19 Epidemic: An Updated Review. Curr Microbiol 2023; 80:322. [PMID: 37592083 DOI: 10.1007/s00284-023-03430-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 07/26/2023] [Indexed: 08/19/2023]
Abstract
Although mucormycosis may have reached an epidemic situation during the COVID-19 pandemic, the term was much more familiar even before the COVID-19 period. The year 2020 showed an outbreak of novel coronavirus (SARS-CoV-2) which affected millions of people all over the world. One of the noticeable complications observed to be associated with this disease is mucormycosis. It is an opportunistic infection caused by members of the Order Mucorales existing worldwide and has been commonly reported as a laboratory contaminant for a long time. However, nowadays due to the changes in the host environment, they have been emerging as potent opportunistic pathogens responsible for causing primary infections or coinfections with other diseases eventually resulting in morbidity and even mortality in severe cases. Although immunocompromised patients are more susceptible to this infection, few cases have been reported in immunocompetent individuals. Various risk factors which are responsible for the acquisition of mucormycosis include diabetes mellitus type 2, ketoacidosis, hematological malignancies, organ transplants, and chemotherapy recipients. Among the various etiological agents, Rhizopus is found to be the most common, and rhino-cerebral to be the most frequent clinical presentation. As far as pathogenesis is concerned, host cell invasion, thrombosis, and necrosis are the main events in the progression of this disease. The aim of the present review is to address a complete spectrum of mucormycosis and COVID-19-associated mucormycosis (CAM) in a single article. Both global and Indian scenarios of mucormycosis are taken into account while framing this review.
Collapse
Affiliation(s)
- Bharti Sharma
- Department of Botany, University of Jammu, Jammu, Jammu and Kashmir, 180006, India
| | - Skarma Nonzom
- Department of Botany, University of Jammu, Jammu, Jammu and Kashmir, 180006, India.
| |
Collapse
|
7
|
Zhou L, Li M, Li H, Guo Z, Gao Y, Zhang H, Qin F, Sang Z, Xing Q, Cheng L, Cao W. Establishment of a mathematical prediction model for voriconazole stable maintenance dose: a prospective study. Front Cell Infect Microbiol 2023; 13:1157944. [PMID: 37565064 PMCID: PMC10410275 DOI: 10.3389/fcimb.2023.1157944] [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: 02/09/2023] [Accepted: 07/11/2023] [Indexed: 08/12/2023] Open
Abstract
Background In patients with invasive fungal infection (IFI), the steady-state serum trough concentration (C min) of voriconazole (VCZ) is highly variable and can lead to treatment failure (C min < 0.5 mg/L) and toxicity (C min ≥ 5.0 mg/L). However, It remains challenging to determine the ideal maintenance dose to achieve the desired C min level quickly. Aims This randomized, prospective observational single-center study aimed to identify factors affecting VCZ-C min and maintenance dose and create an algorithmic model to predict the necessary maintenance dose. MeThe study enrolled 306 adult IFI patients, split into two groups: non-gene-directed (A) (where CYP2C19 phenotype is not involved in determining VCZ dose) and gene-directed (B) (where CYP2C19 phenotype is involved in determining VCZ dose). Results Results indicated that CYP2C19 genetic polymorphisms might significantly impact VCZ loading and maintenance dose selection. CYP2C19 phenotype, C-reaction protein (CRP), and average daily dose/body weight were significant influencers on VCZ-C min, while CYP2C19 phenotype, CRP, and body weight significantly impacted VCZ maintenance dose. A feasible predictive formula for VCZ stable maintenance dose was derived from the regression equation as a maintenance dose (mg) =282.774-0.735×age (year)+2.946×body weight(Kg)-19.402×CYP2C19 phenotype (UM/RM/NM:0, IM:1, PM:2)-0.316×CRP (mg/L) (p < 0.001). Discussion DiThis formula may serve as a valuable supplement to the Clinical Pharmacogenetics Implementation Consortium (CPIC®) guideline for CYP2C19 and VCZ therapy, especially for IFI patients with highly variable inflammatory cytokines during VCZ therapy.
Collapse
Affiliation(s)
- Lijuan Zhou
- Translational Medicine Center, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, Henan, China
| | - Min Li
- Translational Medicine Center, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, Henan, China
| | - Huihong Li
- Translational Medicine Center, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, Henan, China
| | - Zhiqiang Guo
- Department of Hematology, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, Henan, China
| | - Yanqiu Gao
- Department of Respiratory Medicine, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, Henan, China
| | - Hua Zhang
- Department of Respiratory Medicine, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, Henan, China
| | - Fuli Qin
- Department of Hematology, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, Henan, China
| | - Zhihui Sang
- Translational Medicine Center, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, Henan, China
- College of Pharmacy, Xinxiang Medical University, Xinxiang, Henan, China
| | - Qinghe Xing
- Institutes of Biomedical Sciences and Children’s Hospital, Fudan University, Shanghai, China
| | - Long Cheng
- College of Nursing, Chifeng University, Chifeng, Inner Mongolia, China
| | - Wei Cao
- Translational Medicine Center, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
8
|
Ghadimi F, Rodrigues CF, Mohammadi SR, Roudbary M, Dos Santos AL, Aslani P, Nikoomanesh F. Oral candidiasis in patients with kidney transplantation in Iran: prevalence and antifungal susceptibility pattern. Future Microbiol 2023; 18:715-722. [PMID: 37665236 DOI: 10.2217/fmb-2022-0179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/05/2023] Open
Abstract
Aim: This study aimed to identify Candida species recovered from the oral cavity of patients with kidney transplantation. Materials & methods: Two swabs were taken from the oral cavities of 40 patients before and after transplantation, cultured on Sabouraud dextrose agar, and yeasts identified. Antifungal drug susceptibility testing was performed with fluconazole and itraconazole. Results: Candida glabrata was the most frequently isolated species in patients, followed by Candida albicans and Rhodotorula. C. glabrata isolates from patients before transplantation were resistant to fluconazole, whereas C. albicans was fluconazole-resistant both before and after transplantation. Conclusion: The importance of non-albicans Candida species in the oral cavity of patients sheds light on performing antifungal tests for achieving the best outcome to prevent therapeutic failure.
Collapse
Affiliation(s)
- Fardad Ghadimi
- Department of Mycology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, 14115111, Iran
| | - Célia F Rodrigues
- TOXRUN - Toxicology Research Unit, Cooperativa de Ensino Superior Politécnico e Universitário - CESPU, 4585-116 Gandra PRD, Portugal
- LEPABE - Laboratory for Process Engineering, Environment, Biotechnology & Energy, Faculty of Engineering, University of Porto, Rua Dr Roberto Frias, 4200-465 Porto, Portugal
- ALiCE - Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr Roberto Frias, 4200-465 Porto, Portugal
| | - Shahla Roudbar Mohammadi
- Department of Mycology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, 14115111, Iran
| | - Maryam Roudbary
- Department of Parasitology & Mycology, School of Medicine, Iran University of Medical Sciences, Tehran, 1449614535, Iran
- Sydney Institute for Infectious Diseases, University of Sydney, Sydney, NSW2145, Australia
| | - André Ls Dos Santos
- Department of General Microbiology, Microbiology Institute, Federal University of Rio de Janeiro, 21941901, Brazil
| | - Peyman Aslani
- Department of Parasitology & Mycology, Faculty of Medicine, Aja University of Medical Sciences, Tehran, 1411718541, Iran
| | - Fatemeh Nikoomanesh
- Infectious Disease Research Center, Birjand University of Medical Sciences, Birjand, 9717853577, Iran
| |
Collapse
|
9
|
Henning MAS, Hay R, Rodriguez-Cerdeira C, Szepietowski JC, Piraccini BM, Ferreirós MP, Arabatzis M, Sergeev A, Nenoff P, Kotrekhova L, Nowicki RJ, Faergemann J, Padovese V, Prohic A, Skerlev M, Schmid-Grendelmeier P, Sigurgeirsson B, Gaitanis G, Lecerf P, Saunte DML. Position statement: Recommendations on the diagnosis and treatment of Malassezia folliculitis. J Eur Acad Dermatol Venereol 2023. [PMID: 36912427 DOI: 10.1111/jdv.18982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 02/07/2023] [Indexed: 03/14/2023]
Abstract
Malassezia is a lipophilic yeast that is a part of the human mycobiome. Malassezia folliculitis appears when the benign colonization of the hair follicles, by the Malassezia yeasts, becomes symptomatic with pruritic papules and pustules. Although Malassezia folliculitis is common in hospital departments, diagnosing and treating it varies among dermatologists and countries. The European Academy of Dermatology and Venereology Mycology Task Force Malassezia folliculitis working group has, therefore, sought to develop these recommendations for the diagnosis and management of Malassezia folliculitis. Recommendations comprise methods for diagnosing Malassezia folliculitis, required positive findings before starting therapies and specific treatment algorithms for individuals who are immunocompetent, immunocompromised or who have compromised liver function. In conclusion, this study provides a clinical strategy for diagnosing and managing Malassezia folliculitis.
Collapse
Affiliation(s)
- M A S Henning
- Department of Dermatology, Zealand University Hospital, Roskilde, Denmark.,Department of Clinical Medicine, Health Sciences Faculty, University of Copenhagen, Copenhagen, Denmark
| | - R Hay
- King's College London, London, UK
| | | | - J C Szepietowski
- Department of Dermatology, Venereology and Allergology, Wroclaw Medical University, Wroclaw, Poland
| | - B M Piraccini
- Dermatology, IRCCS Policlinico di Sant'Orsola, Bologna, Italy.,Department of Experimental, Diagnostic and Specialty Medicine (DIMES), Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - M P Ferreirós
- Department of Dermatology, Faculty of Medicine, University of Santiago de Compostela, A Coruña, Spain
| | - M Arabatzis
- Dermatology Department, Medical School, University of Thessaloniki, Thessaloniki, Greece
| | - A Sergeev
- All-Russian National Academy of Mycology, Moscow, Russia
| | - P Nenoff
- Laboratory of Medical Microbiology, Mölbis, Germany
| | - L Kotrekhova
- Department of Dermatovenereology, North Western State Medical University, Saint Petersburg, Russia
| | - R J Nowicki
- Department of Dermatology, Venereology, and Allergology, Medical University of Gdansk, Gdańsk, Poland
| | - J Faergemann
- Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - V Padovese
- Department of Dermatology and Venereology, Genitourinary Clinic, Mater Dei Hospital, Msida, Malta.,International Foundation for Dermatology, Migrants Health Dermatology Working Group, London, UK
| | - A Prohic
- Department of Dermatovenereology, University Sarajevo School of Science and Technology, Sarajevo Medical School, Sarajevo, Bosnia and Herzegovina
| | - M Skerlev
- Department of Dermatology and Venereology, Zagreb University School of Medicine and Zagreb University Hospital, Zagreb, Croatia
| | - P Schmid-Grendelmeier
- Allergy Unit, Department of Dermatology, University Hospital Zurich and Medical Faculty, University of Zurich, Zurich, Switzerland
| | - B Sigurgeirsson
- Department of Dermatology, Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - G Gaitanis
- Department of Skin and Venereal Diseases, Faculty of Medicine, School of Health Sciences, University of Ioannina, Ioannina, Greece
| | - P Lecerf
- Department of Dermatology, University Hospital Brugmann & Saint-Pierre, Université Libre de Bruxelles, Brussels, Belgium
| | - D M L Saunte
- Department of Dermatology, Zealand University Hospital, Roskilde, Denmark.,Department of Clinical Medicine, Health Sciences Faculty, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
10
|
In Silico and In Vitro Analysis of Sulforaphane Anti- Candida Activity. Antibiotics (Basel) 2022; 11:antibiotics11121842. [PMID: 36551499 PMCID: PMC9774275 DOI: 10.3390/antibiotics11121842] [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: 10/31/2022] [Revised: 12/07/2022] [Accepted: 12/15/2022] [Indexed: 12/23/2022] Open
Abstract
Oropharyngeal candidiasis/candidosis is a common and recurrent opportunistic fungal infection. Fluconazole (FLZ), one of the most used and effective antifungal agents, has been associated with a rise of resistant Candida species in immunocompromised patients undergoing prophylactic therapy. Sulforaphane (SFN), a compound from cruciferous vegetables, is an antimicrobial with yet controversial activities and mechanisms on fungi. Herein, the in silico and antifungal activities of SFN against C. albicans were investigated. In silico analyzes for the prediction of the biological activities and oral bioavailability of SFN, its possible toxicity and pharmacokinetic parameters, as well as the estimates of its gastrointestinal absorption, permeability to the blood-brain barrier and skin, and similarities to drugs, were performed by using different software. SFN in vitro anti-Candida activities alone and in combination with fluconazole (FLZ) were determined by the broth microdilution method and the checkerboard, biofilm and hyphae formation tests. Amongst the identified probable biological activities of SFN, nine indicated an antimicrobial potential. SFN was predicted to be highly absorbable by the gastrointestinal tract, to present good oral availability, and not to be irritant and/or hepatotoxic. SFN presented antifungal activity against C. albicans and prevented both biofilm and hyphae formation by this microorganism. SFN was additive/synergistic to FLZ. Overall, the data highlights the anti-Candida activity of SFN and its potential to be used as an adjuvant therapy to FLZ in clinical settings.
Collapse
|
11
|
Voznesenskiy S, Ermak T, Emerole K, Samotolkina Е, Klimkova P, Abramova E, Kozhevnikova G. Coexistence of Competing Opportunistic Pathogens in Critically ill Patients with Advanced AIDS: A Case Report and Literature Review. Open AIDS J 2022. [DOI: 10.2174/18746136-v16-e2208040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Introduction:
Opportunistic infections (OIs) are the leading causes of morbidity and mortality among HIV-infected individuals. The incidence of OIs is greater in antiretroviral treatment (ART) naive patients. As of 30 June 2021, 28.2 (73%) people with HIV/AIDS (PLWHA) were accessing antiretroviral therapy (ART) globally, leaving the remaining 27% PLWHA without ART at risk for OIs. Multiple opportunistic infections are caused due to the coexistence of competing opportunistic pathogens that confound clinical manifestations, investigative procedures, and management protocols.
Case Presentation:
In this report, we describe the case of a critically ill HIV female patient admitted to the ICU. The patient was diagnosed with multiple opportunistic infections and subsequently died after her illness progressed. Due to the paucity of information on the subject, we conducted a retrospective study of 1440 case records of HIV/AIDS critically ill patients to determine the incidence and spectrum of multiple opportunistic infections. We performed a review of the available medical literature relevant to the subject.
Conclusion:
Knowledge of such events would guide and enhance the physician's diagnostic and management strategies, especially in resource limited regions.
Collapse
|
12
|
Mwassi HA, Yahav D, Ayada G, Matsri S, Margalit I, Shargian L, Bishara J, Atamna A. Systemic anti-fungal therapy for esophageal candidiasis – systematic review and meta-analysis of randomized controlled trials. Int J Antimicrob Agents 2022; 59:106590. [DOI: 10.1016/j.ijantimicag.2022.106590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Revised: 03/25/2022] [Accepted: 04/03/2022] [Indexed: 11/16/2022]
|
13
|
Peng Z, Tang J. Intestinal Infection of Candida albicans: Preventing the Formation of Biofilm by C. albicans and Protecting the Intestinal Epithelial Barrier. Front Microbiol 2022; 12:783010. [PMID: 35185813 PMCID: PMC8847744 DOI: 10.3389/fmicb.2021.783010] [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: 09/25/2021] [Accepted: 12/30/2021] [Indexed: 12/12/2022] Open
Abstract
The large mortality and morbidity rate of C. albicans infections is a crucial problem in medical mycology. Because the generation of biofilms and drug resistance are growing concerns, the growth of novel antifungal agents and the looking for newer objectives are necessary. In this review, inhibitors of C. albicans biofilm generation and molecular mechanisms of intestinal epithelial barrier protection are elucidated. Recent studies on various transcription elements; quorum-sensing molecules; host responses to adherence; and changes in efflux pumps, enzymes, bud to hyphal transition, and lipid profiles have increased the knowledge of the intricate mechanisms underlying biofilm resistance. In addition, the growth of novel biomaterials with anti-adhesive nature, natural products, drugs, bioactive compounds, proteins, lipids, and carbohydrates are being researched. Recently, more and more attention has been given to various metal nanoparticles that have also appeared as antibiofilm agents in C. albicans. The intestinal epithelial obstacle exerts an crucial effect on keeping intestinal homeostasis and is increasingly associated with various disorders associated with the intestine such as inflammatory bowel disease (IBD), irritable bowel syndrome, metabolic syndrome, allergies, hepatic inflammation, septic shock, etc. However, whether their involvement in the prevention of other intestinal disorders like IBD are useful in C. albicans remains unknown. Further studies must be carried out in order to validate their inhibition functions in intestinal C. albicans. This provides innovates ideas for intestinal C. albicans treatment.
Collapse
Affiliation(s)
- Ziyao Peng
- Department of Trauma-Emergency and Critical Care Medicine, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Jianguo Tang
- Department of Trauma-Emergency and Critical Care Medicine, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| |
Collapse
|
14
|
Zeng BS, Zeng BY, Hung CM, Chen TY, Wu YC, Tu YK, Lin PY, Su KP, Stubbs B, Sun CK, Cheng YS, Li DJ, Liang CS, Hsu CW, Chen YW, Tseng PT, Chen CH. Efficacy and acceptability of different anti-fungal interventions in oropharyngeal or esophageal candidiasis in HIV co-infected adults: a pilot network meta-analysis. Expert Rev Anti Infect Ther 2021; 19:1469-1479. [PMID: 33899657 DOI: 10.1080/14787210.2021.1922078] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Background: Oropharyngeal/esophageal candidiasis are the most common opportunistic infections observed in patients with human immunodeficiency virus (HIV). While the commonly recommended treatment is fluconazole, relapse of oropharyngeal or esophageal candidiasis has been gradually increasing in recent decades.Methods: The current network meta-analysis (NMA) included randomized controlled trials (RCTs) investigating the efficacy and acceptability (i.e. drop-out rate) of different anti-fungal interventions against oropharyngeal or esophageal candidiasis in adults with HIV. All NMA procedures were conducted using the frequentist model.Results: Twenty-seven RCTs and 6277 participants were included. For oropharyngeal candidiasis, photosensitizer-based antimicrobial photodynamic therapy (aPDT) with laser irradiation plus methylene blue was associated with the highest cure rate and the lowest relapse rate among the investigated interventions [odds ratio (OR) = 6.82, 95% confidence intervals (95%CIs) = 0.19 to 244.42, p = 0.293, and OR = 0.03, 95%CIs = 0.00 to 0.77, p = 0.034, compared to fluconazole]. None of the investigated anti-fungal interventions were superior to fluconazole for esophageal candidiasis in respect of cure rates/relapse rates. All investigated anti-fungal interventions were well-accepted.Conclusions: aPDT could be the preferred strategy to manage oropharyngeal candidiasis; however the evidence for esophageal candidiasis still remained inconclusive.
Collapse
Affiliation(s)
- Bing-Syuan Zeng
- Department of Internal Medicine, E-Da Hospital, Kaohsiung, Taiwan
| | - Bing-Yan Zeng
- Department of Internal Medicine, E-Da Hospital, Kaohsiung, Taiwan
| | - Chao-Ming Hung
- Division of General Surgery, Department of Surgery, E-Da Cancer Hospital, Kaohsiung, Taiwan.,School of Medicine, College of Medicine, I-Shou University, Kaohsiung, Taiwan
| | - Tien-Yu Chen
- Department of Psychiatry, Tri-Service General Hospital; School of Medicine, National Defense Medical Center, Taipei, Taiwan.,Institute of Brain Science, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yi-Cheng Wu
- Department of Sports Medicine, Landseed International Hospital, Taoyuan, Taiwan
| | - Yu-Kang Tu
- Institute of Epidemiology & Preventive Medicine, College of Public Health, National Taiwan University, Taipei, Taiwan.,Department of Dentistry, National Taiwan University Hospital, Taipei, Taiwan
| | - Pao-Yen Lin
- Department of Psychiatry, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan.,Institute for Translational Research in Biomedical Sciences, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Kuan-Pin Su
- Department of Psychiatry & Mind-Body Interface Laboratory (Mbi-lab), China Medical University Hospital, Taichung, Taiwan.,College of Medicine, China Medical University, Taichung, Taiwan.,An-Nan Hospital, China Medical University, Tainan, Taiwan.,Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Brendon Stubbs
- Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK.,Physiotherapy Department, South London and Maudsley NHS Foundation Trust, London, UK.,Faculty of Health, Social Care Medicine and Education, Anglia Ruskin University, Chelmsford, UK
| | - Cheuk-Kwan Sun
- Department of Emergency Medicine, E-Da Hospital, I-Shou University School of Medicine for International Students, Kaohsiung, Taiwan
| | - Yu-Shian Cheng
- Department of Psychiatry, Tsyr-Huey Mental Hospital, Kaohsiung, Taiwan
| | - Dian-Jeng Li
- Department of Addiction Science, Kaohsiung Municipal Kai-Syuan Psychiatric Hospital, Kaohsiung City, Taiwan
| | - Chih-Sung Liang
- Department of Psychiatry, Beitou Branch, Tri-Service General Hospital; School of Medicine, National Defense Medical Center, Taipei, Taiwan.,Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Chih-Wei Hsu
- Department of Psychiatry, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Yen-Wen Chen
- Prospect Clinic for Otorhinolaryngology & Neurology, Kaohsiung, Taiwan
| | - Ping-Tao Tseng
- Prospect Clinic for Otorhinolaryngology & Neurology, Kaohsiung, Taiwan.,Institute of Biomedical Sciences, National Sun Yat-sen University, Kaohsiung, Taiwan.,Department of Psychology, College of Medical and Health Science, Asia University, Taichung, Taiwan
| | - Chang-Hua Chen
- Program in Translational Medicine, National Chung Hsing University, Taichung City, Taiwan.,Rong Hsing Research Center For Translational Medicine, National Chung Hsing University, Taichung City, Taiwan.,Department of Internal Medicine, Changhua Christian Hospital, Changhua, Taiwan
| |
Collapse
|
15
|
Favorable Effects of Voriconazole Trough Concentrations Exceeding 1 μg/mL on Treatment Success and All-Cause Mortality: A Systematic Review and Meta-Analysis. J Fungi (Basel) 2021; 7:jof7040306. [PMID: 33923727 PMCID: PMC8072959 DOI: 10.3390/jof7040306] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 04/14/2021] [Accepted: 04/15/2021] [Indexed: 12/23/2022] Open
Abstract
This systematic review and meta-analysis examined the optimal trough concentration of voriconazole for adult patients with invasive fungal infections. We used stepwise cutoffs of 0.5-2.0 μg/mL for efficacy and 3.0-6.0 μg/mL for safety. Studies were included if they reported the rates of all-cause mortality and/or treatment success, hepatotoxicity, and nephrotoxicity according to the trough concentration. Twenty-five studies involving 2554 patients were included. The probability of mortality was significantly decreased using a cutoff of ≥1.0 μg/mL (odds ratio (OR) = 0.34, 95% confidence interval (CI) = 0.15-0.80). Cutoffs of 0.5 (OR = 3.48, 95% CI = 1.45-8.34) and 1.0 μg/mL (OR = 3.35, 95% CI = 1.52-7.38) also increased the treatment success rate. Concerning safety, significantly higher risks of hepatotoxicity and neurotoxicity were demonstrated at higher concentrations for all cutoffs, and the highest ORs were recorded at 4.0 μg/mL (OR = 7.39, 95% CI = 3.81-14.36; OR = 5.76, 95% CI 3.14-10.57, respectively). Although further high-quality trials are needed, our findings suggest that the proper trough concentration for increasing clinical success while minimizing toxicity is 1.0-4.0 μg/mL for adult patients receiving voriconazole therapy.
Collapse
|
16
|
Abstract
Among all the viral infections, acquired immunodeficiency syndrome (AIDS) is considered as one of the most morbid infections caused by the human immunodeficiency virus (HIV). The prime reason for the pathogenesis is the profound immunosuppression that leads to lethal opportunistic infections (OI), neurological disorders, unexpected malignancies and pathologies of the orofacial region. Patients with OI whose HIV status is unknown have shown a mortality rate higher than those with known HIV status. Among HIV-associated infections, orofacial lesions contribute a major proportion of the OI attributed to the plethora of micro-organisms present in the oral cavity. Apart from serious clinical manifestations, opportunistic infections also lead to significant impairment of quality of life. These lesions not only indicate the HIV infection but also among the clinical manifestations, which often occur early in the course of disease. World Health Organization has also provided policies for treatment/prevention of oral lesions, strengthening the promotion and care of oral health in HIV/AIDS patients. The present review provides comprehensive information about orofacial OI in HIV/AIDS patients and emphasis was also given to the malignancies associated with EB and HTLV virus.
Collapse
|
17
|
Nast A, Smith C, Spuls P, Avila Valle G, Bata‐Csörgö Z, Boonen H, De Jong E, Garcia‐Doval I, Gisondi P, Kaur‐Knudsen D, Mahil S, Mälkönen T, Maul J, Mburu S, Mrowietz U, Reich K, Remenyik E, Rønholt K, Sator P, Schmitt‐Egenolf M, Sikora M, Strömer K, Sundnes O, Trigos D, Van Der Kraaij G, Yawalkar N, Dressler C. EuroGuiDerm Guideline on the systemic treatment of Psoriasis vulgaris – Part 1: treatment and monitoring recommendations. J Eur Acad Dermatol Venereol 2020; 34:2461-2498. [DOI: 10.1111/jdv.16915] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 08/13/2020] [Indexed: 12/14/2022]
Affiliation(s)
- A. Nast
- Department of Dermatology, Venereology and Allergology Charité – Universitätsmedizin BerlinCorporate Member of Freie Universität BerlinHumboldt‐Universität zu BerlinBerlin Institute of Health Berlin Germany
| | - C. Smith
- St John’s Institute of Dermatology London UK
| | - P.I. Spuls
- Academic Medical Centre Amsterdam Amsterdam Netherlands
| | - G. Avila Valle
- Department of Dermatology, Venereology and Allergology Charité – Universitätsmedizin BerlinCorporate Member of Freie Universität BerlinHumboldt‐Universität zu BerlinBerlin Institute of Health Berlin Germany
| | | | - H. Boonen
- Office‐Based Dermatology Practice Geel Belgium
| | - E. De Jong
- Radboud University medical centre Nijmegen Netherlands
| | - I. Garcia‐Doval
- Unidad de InvestigaciónFundación Piel Sana AEDV Madrid España
| | | | | | - S. Mahil
- Guy's and St Thomas' NHS Foundation Trust London UK
| | - T. Mälkönen
- Helsinki University Central Hospital Helsinki Finland
| | - J.T. Maul
- Department of Dermatology University Hospital of Zürich Zürich Switzerland
| | - S. Mburu
- International Federation of Psoriasis Associations (IFPA)
| | - U. Mrowietz
- Universitätsklinikum Schleswig‐Holstein Kiel Germany
| | - K. Reich
- Translational Research in Inflammatory Skin Diseases Institute for Health Services Research in Dermatology and Nursing University Medical Center Hamburg‐Eppendorf Hamburg Germany
| | | | | | - P.G. Sator
- Municipal Hospital Hietzing Vienna Austria
| | - M. Schmitt‐Egenolf
- Dermatology Department of Public Health & Clinical Medicine Umeå University Umeå Sweden
| | - M. Sikora
- Department of Dermatology Medical University of Warsaw Warszawa Poland
| | - K. Strömer
- Office‐Based Dermatology Practice Mönchengladbach Germany
| | | | - D. Trigos
- International Federation of Psoriasis Associations (IFPA)
| | | | - N. Yawalkar
- Department of Dermatology, Inselspital Bern University HospitalUniversity of Bern Bern Switzerland
| | - C. Dressler
- Department of Dermatology, Venereology and Allergology Charité – Universitätsmedizin BerlinCorporate Member of Freie Universität BerlinHumboldt‐Universität zu BerlinBerlin Institute of Health Berlin Germany
| |
Collapse
|
18
|
Dockrell DH, O’Shea D, Cartledge JD, Freedman AR. British HIV Association guidelines on the management of opportunistic infection in people living with HIV: The clinical management of Candidiasis 2019. HIV Med 2020; 20 Suppl 8:2-24. [PMID: 31670458 DOI: 10.1111/hiv.12806] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- D H Dockrell
- University of Edinburgh, Edinburgh, UK and Regional Infectious Diseases Unit, NHS Lothian Infection Service, Edinburgh, UK
| | - D O’Shea
- University of Edinburgh, Edinburgh, UK and Regional Infectious Diseases Unit, NHS Lothian Infection Service, Edinburgh, UK
| | | | - A R Freedman
- Cardiff University School of Medicine, Cardiff, UK
| |
Collapse
|
19
|
Nosratzehi T, Nosratzehi M, Nosratzehi S, Lotfi F. The comparison of the effect of curcumin with nystatin on inhibition level of Candida albicans. J Exp Pharmacol 2019; 11:93-97. [PMID: 31496837 PMCID: PMC6701643 DOI: 10.2147/jep.s215843] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 07/11/2019] [Indexed: 11/23/2022] Open
Abstract
Background Curcumin is a kind of medicinal plant, a member of the ginger family, Zingiberaceae. As the herbal medicine has been globally demanded in recent years and due to the anti-viral, anti-inflammatory, and antifungal properties of curcumin, the present study aimed to compare the inhibitory effect of curcumin with that of nystatin on Candida albicans growth. Methods In this experimental study, the antifungal effect of curcumin on Candida albicans was evaluated. Candida albicans is cultivated in Agar Sabuard medium containing chlorophenic, 10-series of curcumin extract, and one 10-series of a disc impregnated with nystatin. Inside of each plate, one disc of herbal extract, one disc of nystatin as a positive control, one disc of methanol and one empty disc as negative control were placed, after 24 hrs, the inhibitory zone diameter of the herbal extract was compared with that of positive control using Mann–Whitney. Results The mean of inhibitory zone diameter in nystatin and curcumin was 1.04±20.46 and 1.36±0.89 mm, respectively. Mann–Whitney test shows a significant difference between the inhibitory zone diameter of these materials (P<0.001). Conclusion The results of this study suggested that curcumin extract did not have any effect on the inhibition of Candida albicans in laboratory environment.
Collapse
Affiliation(s)
- Tahereh Nosratzehi
- Dental Research Center and Department of Oral Medicine, School of Dentistry, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Mahin Nosratzehi
- Department of Endocrinology, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Shahin Nosratzehi
- Department of Endocrinology and Metabolism Fellowship, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Lotfi
- School of Dentistry, Zahedan University of Medical Sciences, Zahedan, Iran
| |
Collapse
|
20
|
Epidemiology and antifungal susceptibility testing of non-albicansCandida species colonizing mucosae of HIV-infected patients in Yaoundé (Cameroon). J Mycol Med 2019; 29:233-238. [PMID: 31204235 DOI: 10.1016/j.mycmed.2019.06.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 06/04/2019] [Accepted: 06/04/2019] [Indexed: 12/17/2022]
Abstract
Non-albicans Candida (NAC) species have emerged as potent pathogenic yeasts among HIV-infected patients. Authors evaluated the epidemiology and antifungal susceptibility testing of non-albicansCandida species colonizing Yaoundé (capital of the Republic of Cameroon, Central Africa) HIV-infected patients. The mucosal specimens were collected and submitted to the mycological diagnosis. Yeast isolates were identified by the Matrix Assisted Laser Desorption Ionisation - Time of Flight Mass Spectrometry (MALDI-TOF MS). The antifungal susceptibility testing was achieved by the CLSI-M27 protocols, and the interpretation of clinical break points (CBPs) and epidemiological cutoff values were in accordance with the CLSI-M60 and M59 recommendations. Four hundred and two patients were recruited and 1218 samples collected. The colonisation frequency was 24.1% and 304 yeasts isolated. Yeast isolates were 113 (37.2%) C. albicans, 2 (0.7%) C. africana and 172 (56.6%) NAC isolates. The NAC isolates were grouped into 13 species including C. krusei (18.1%), C. glabrata (10.9%), C. tropicalis (8.5%) and C. parapsilosis (5.9%) as the major ones. All the isolates appeared to be wild-type for amphotericin B and itraconazole. One (1/33) isolate of C. glabrata was resistant to fluconazole. C. arapsilosis isolates appeared all susceptible to fluconazole. C. tropicalis isolates presented 50% (13/26) resistance to fluconazole. The achieved results bring out new insights about epidemiology of NAC species in Cameroon. The results also highlight the resistance of NAC species to current antifungal drugs.
Collapse
|
21
|
Ghrenassia E, Mokart D, Mayaux J, Demoule A, Rezine I, Kerhuel L, Calvet L, De Jong A, Azoulay E, Darmon M. Candidemia in critically ill immunocompromised patients: report of a retrospective multicenter cohort study. Ann Intensive Care 2019; 9:62. [PMID: 31161475 PMCID: PMC6546779 DOI: 10.1186/s13613-019-0539-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2019] [Accepted: 05/26/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Immunocompromised critically ill patients constitute a population with the high risk of candidemia. This retrospective study aimed to assess the outcome of immunocompromised critically ill patients with candidemia. Secondary objectives were to describe clinical phenotypes of these patients, Candida ecology, and factors associated with mortality. RESULTS Overall, 121 patients were included in this study. Median delay from candidemia to first antifungal therapy was 3 days, in line with the observed delay of blood culture positivity. Candia albicans was the main Candida specie identified (54%), and susceptibility of Candida to fluconazole and echinocandins was of, respectively, 70% and 92%. Hospital mortality was of 60%. After adjustment for confounders, severity as assessed by the need for vasopressors (HR 1.8, CI95% 1.1-3.1), need for mechanical ventilation (HR 2.0, CI95% 1.1-3.8) and allogenic stem cell transplantation (HR 2.5, CI95% 1.1-6.0) were independently associated with poor outcome. Candida specie, susceptibility and treatment strategies were not associated with outcome. CONCLUSIONS Candidemia in immunocompromised critically ill patients is associated with a grim outcome. Despite the high prevalence of Candida non-albicans species, neither C. species nor its susceptibility was associated with outcome. Conversely, severity and preexisting allogeneic stem cell transplantation were independently associated with poor outcome. Despite antifungal prophylaxis and use of preemptive antifungal therapy in neutropenic patients, antifungal therapy was initiated three days after symptoms onset suggesting needs for specific strategies aiming to reduce this delay.
Collapse
Affiliation(s)
- Etienne Ghrenassia
- Medical ICU, Saint Louis Hospital, Paris, France
- Paris Diderot University, Paris, France
- Sorbonne University, Paris, France
| | | | - Julien Mayaux
- Medical ICU, Pitié-Salpétrière Hospital, Paris, France
| | - Alexandre Demoule
- Sorbonne University, Paris, France
- Medical ICU, Pitié-Salpétrière Hospital, Paris, France
| | - Imène Rezine
- Medical ICU, Saint Louis Hospital, Paris, France
| | - Lionel Kerhuel
- Medical ICU, Saint Louis Hospital, Paris, France
- Paris Diderot University, Paris, France
| | - Laure Calvet
- Medical ICU, Saint Louis Hospital, Paris, France
| | - Audrey De Jong
- Medical ICU, Saint Louis Hospital, Paris, France
- Département d’anesthésie-réanimation Saint-Eloi, PhyMedExp, INSERM, CNRS, CHU de Montpellier, Université de Montpellier, 80, avenue Augustin-Fliche, 34295 Montpellier Cedex, France
| | - Elie Azoulay
- Medical ICU, Saint Louis Hospital, Paris, France
- Paris Diderot University, Paris, France
| | - Michael Darmon
- Medical ICU, Saint Louis Hospital, Paris, France
- Paris Diderot University, Paris, France
- ECSTRA Team, Biostatistics and Clinical Epidemiology, UMR 1153 (Center of Epidemiology and Biostatistic Sorbonne Paris Cité, CRESS), INSERM, Paris, France
- Public Health Department, Robert Debré University Hospital, AP-HP, Paris, France
| |
Collapse
|
22
|
Zhong X, Tong X, Ju Y, Du X, Li Y. Interpersonal Factors in the Pharmacokinetics and Pharmacodynamics of Voriconazole: Are CYP2C19 Genotypes Enough for Us to Make a Clinical Decision? Curr Drug Metab 2019; 19:1152-1158. [PMID: 29361899 PMCID: PMC6635675 DOI: 10.2174/1389200219666171227200547] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 09/04/2017] [Accepted: 11/26/2017] [Indexed: 02/08/2023]
Abstract
Background: Invasive mycoses are serious infections with high mortality and increasing inci-dence. Voriconazole, an important drug to treat invasive mycosis, is metabolized mainly by the cytochrome P450 family 2 subfamily C member 19 enzyme (CYP2C19) and is affected by the genotypes of CYP2C19. Objective: We reviewed studies on how genotypes affect the pharmacokinetics and pharmacodynamics of voriconazole, and attempted to determine a method to decide on dosage adjustments based on genotypes, after which, the main characteristic of voriconazole was clarified in details. The pharmacokinetics of voriconazole are influenced by various inter and intrapersonal factors, and for certain populations, such as geriatric patients and pediatric patients, these influences must be considered. CYP2C19 genotype represents the main part of the interpersonal variability related to voriconazole blood concentrations. Thus monitoring the concentration of voriconazole is needed in clinical scenarios to minimize the negative influences of inter and intrapersonal factors. Several studies provided evidence on the stable trough concentration range from 1-2 to 4-6 mg/L, which was combined to consider the efficacy and toxicity. However, the therapeutic drug concentration needs to be narrowed down and evaluated by large-scale clinical trials. Conclusion: Though there is insufficient evidence on the relationship between CYP2C19 genotypes and clinical outcomes, there is a great potential for the initial voriconazole dose selection to be guided by the CYP2C19 genotype. Finally, voriconazole therapeutic drug monitoring is essential to provide patient-specific dosing recommendations, leading to more effective anti-fungal regimens to increase clinical effica-cy and reduce adverse drug reactions.
Collapse
Affiliation(s)
- Xuefeng Zhong
- Department of Respiratory and Critical Care Medicine, National Center of Gerontology, Beijing Hospital, National Clinical Research Center for Respiratory Diseases, Beijing, China
| | - Xunliang Tong
- Department of Geriatrics, Beijing Hospital, National Center of Gerontology, Beijing, China
| | - Yang Ju
- Department of Respiratory and Critical Care Medicine, National Center of Gerontology, Beijing Hospital, National Clinical Research Center for Respiratory Diseases, Beijing, China
| | - Xiaoman Du
- Department of Respiratory and Critical Care Medicine, National Center of Gerontology, Beijing Hospital, National Clinical Research Center for Respiratory Diseases, Beijing, China
| | - Yanming Li
- Department of Respiratory and Critical Care Medicine, National Center of Gerontology, Beijing Hospital, National Clinical Research Center for Respiratory Diseases, Beijing, China
| |
Collapse
|
23
|
von Lilienfeld-Toal M, Wagener J, Einsele H, A. Cornely O, Kurzai O. Invasive Fungal Infection. DEUTSCHES ARZTEBLATT INTERNATIONAL 2019; 116:271-278. [PMID: 31159914 PMCID: PMC6549129 DOI: 10.3238/arztebl.2019.0271] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 06/29/2018] [Accepted: 02/14/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND The incidence of invasive fungal infection is approximately 6 cases per 100 000 persons per year. It is estimated that only half of such infections are detected during the patient's lifetime, making this one of the more common overlooked causes of death in intensive-care patients. The low detection rate is due in part to the complexity of the diagnostic work-up, in which the clinical, radiological, and microbiological findings must be considered. Fungi with resistance to antimycotic drugs have been found to be on the rise around the world. METHODS This review is based on pertinent publications retrieved from a selective search in PubMed, with special attention to guidelines on the diagnosis and treatment of invasive fungal infections caused by Candida spp., Aspergillus spp., Mucorales, and Fusarium spp. RESULTS The clinical risk factors for invasive fungal infection include, among others, congenital immune deficiency, protracted (>10 days) marked granulocytopenia (<0.5 x 109/L), allogeneic stem-cell transplantation, and treatment with immunosuppressive drugs or corticosteroids. High-risk groups include patients in intensive care and those with structural pulmonary disease and/or compli- cated influenza. The first line of treatment, supported by the findings of randomized clinical trials, consists of echinocandins for in- fections with Candida spp. (candidemia response rates: 75.6% for anidulafungin vs. 60.2% for fluconazole) and azole antimycotic drugs for infections with Aspergillus spp. (response rates: 52.8% for voriconazole vs. 31.6% for conventional amphotericin B). The recommended first-line treatment also depends on the local epidemiology. This challenge should be met by interdisciplinary collaboration. Therapeutic decision-making should also take account of the often severe undesired effects of antimycotic drugs (including impairment of hepatic and/or renal function) and the numerous interactions that some of them have with other drugs. CONCLUSION Invasive fungal infections are often overlooked in routine hospital care. They should be incorporated into antimicro- bial stewardship programs as an essential component. There is also a pressing need for the development of new classes of antimycotic drug.
Collapse
Affiliation(s)
- Marie von Lilienfeld-Toal
- National Reference Center for Invasive Fungal Infections, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute (HKI), Jena
- Clinic of Internal Medicine II, University Hospital Jena
| | - Johannes Wagener
- National Reference Center for Invasive Fungal Infections, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute (HKI), Jena
- Institute for Hygiene and Microbiology, University of Würzburg, Chair of Medical Microbiology and Mycology, Würzburg
| | - Hermann Einsele
- Department of Internal Medicine II, University Hospital of Würzburg
- InfectControl 2020, Jena/Würzburg
| | - Oliver A. Cornely
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Department I of Internal Medicine, at the University Hospital of Cologne, European Excellence Center for Medical Mycology (ECMM), DGerman Center for Infection Research(DZIF) Partner Site Bonn Köln, Cologne University
| | - Oliver Kurzai
- National Reference Center for Invasive Fungal Infections, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute (HKI), Jena
- Institute for Hygiene and Microbiology, University of Würzburg, Chair of Medical Microbiology and Mycology, Würzburg
- InfectControl 2020, Jena/Würzburg
| |
Collapse
|
24
|
Yu SY, Zhang L, Chen S, Kong F, Xiao M, Wang H, Hou X, Zhou ML, Zhang G, Zhang JJ, Duan SM, Kang W, Xu YC. Candida isolates causing refractory or recurrent oropharyngeal candidiasis in 11 hospitals in China. Infect Drug Resist 2019; 12:865-875. [PMID: 31114266 PMCID: PMC6489570 DOI: 10.2147/idr.s199359] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Accepted: 03/19/2019] [Indexed: 12/24/2022] Open
Abstract
Introduction: We studied the species distribution and antifungal susceptibilities of Candida isolates causing refractory or recurrent oropharyngeal candidiasis (OPC) in a multicenter study in China (2013–2016). Methods: Species identification was performed using the Bruker Biotyper (Bruker Daltonics, Germany) matrix-assisted laser desorption/ionization time of flight mass spectrometry system supplemented by internal transcribed spacer sequencing as required. Antifungal susceptibilities were determined by the Clinical and Laboratory Standards Institute document (CLSI) M27-A3 broth microdilution methodology. Results: A total of 558 non-duplicate Candida isolates comprising 10 species were obtained from 535 patients. Candida albicans was the most common species (89.6%), followed by C. glabrata (5.2%), C. tropicalis (2.9%), and C. parapsilosis (0.7%). Azoles were active against C. albicans with susceptibility rates of 96% and 95.8% for fluconazole and voriconazole, respectively. MIC50 values of C. albicans to fluconazole, voriconazole, itraconazole, and miconazole were 1, 0.03, 0.25 and 0.12 μg/mL, respectively, higher than those in previous studies of which OPC patients (corresponding MIC50 values of 0.25 , 0.015 , 0.06 , and 0.03 μg/mL). Except for itraconazole, the MIC50 and MIC90 values of 58 non-C. albicans to other azoles were two to threefold higher than C. albicans. Miconazole, amphotericin B, nystatin, and 5-flucytosine had good in vitro antifungal activity for all isolates. Conclusion: The study provides valuable data on the species distribution and antifungal susceptibility of oropharyngeal Candida isolates from geographically diverse areas of China. C. albicans remains the most common species but with increasing rates of azoles resistance.
Collapse
Affiliation(s)
- Shu-Ying Yu
- Department of Clinical Laboratory, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing 100730, People's Republic of China.,Graduate School, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, People's Republic of China.,Beijing Key Laboratory for Mechanisms Research and Precision Diagnosis of Invasive Fungal Diseases (BZ0447), Beijing, People's Republic of China
| | - Li Zhang
- Department of Clinical Laboratory, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing 100730, People's Republic of China.,Beijing Key Laboratory for Mechanisms Research and Precision Diagnosis of Invasive Fungal Diseases (BZ0447), Beijing, People's Republic of China
| | - Sharon Chen
- Centre for Infectious Diseases and Microbiology Laboratory Services, ICPMR -New South Wales Health Pathology, The University of Sydney, Westmead, New South Wales, Australia
| | - Fanrong Kong
- Centre for Infectious Diseases and Microbiology Laboratory Services, ICPMR -New South Wales Health Pathology, The University of Sydney, Westmead, New South Wales, Australia
| | - Meng Xiao
- Department of Clinical Laboratory, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing 100730, People's Republic of China.,Beijing Key Laboratory for Mechanisms Research and Precision Diagnosis of Invasive Fungal Diseases (BZ0447), Beijing, People's Republic of China
| | - He Wang
- Department of Clinical Laboratory, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing 100730, People's Republic of China.,Beijing Key Laboratory for Mechanisms Research and Precision Diagnosis of Invasive Fungal Diseases (BZ0447), Beijing, People's Republic of China
| | - Xin Hou
- Department of Clinical Laboratory, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing 100730, People's Republic of China.,Graduate School, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, People's Republic of China.,Beijing Key Laboratory for Mechanisms Research and Precision Diagnosis of Invasive Fungal Diseases (BZ0447), Beijing, People's Republic of China
| | - Meng-Lan Zhou
- Department of Clinical Laboratory, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing 100730, People's Republic of China.,Graduate School, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, People's Republic of China.,Beijing Key Laboratory for Mechanisms Research and Precision Diagnosis of Invasive Fungal Diseases (BZ0447), Beijing, People's Republic of China
| | - Ge Zhang
- Department of Clinical Laboratory, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing 100730, People's Republic of China.,Beijing Key Laboratory for Mechanisms Research and Precision Diagnosis of Invasive Fungal Diseases (BZ0447), Beijing, People's Republic of China
| | - Jing-Jia Zhang
- Department of Clinical Laboratory, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing 100730, People's Republic of China.,Beijing Key Laboratory for Mechanisms Research and Precision Diagnosis of Invasive Fungal Diseases (BZ0447), Beijing, People's Republic of China
| | - Si-Meng Duan
- Department of Clinical Laboratory, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing 100730, People's Republic of China.,Beijing Key Laboratory for Mechanisms Research and Precision Diagnosis of Invasive Fungal Diseases (BZ0447), Beijing, People's Republic of China
| | - Wei Kang
- Department of Clinical Laboratory, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing 100730, People's Republic of China.,Beijing Key Laboratory for Mechanisms Research and Precision Diagnosis of Invasive Fungal Diseases (BZ0447), Beijing, People's Republic of China
| | - Ying-Chun Xu
- Department of Clinical Laboratory, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing 100730, People's Republic of China.,Beijing Key Laboratory for Mechanisms Research and Precision Diagnosis of Invasive Fungal Diseases (BZ0447), Beijing, People's Republic of China
| |
Collapse
|
25
|
Goemaere B, Lagrou K, Spriet I, Hendrickx M, Vandael E, Becker P, Catry B. Systemic antifungal drug use in Belgium—One of the biggest antifungal consumers in Europe. Mycoses 2019; 62:542-550. [DOI: 10.1111/myc.12912] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2018] [Revised: 03/28/2019] [Accepted: 03/14/2019] [Indexed: 11/26/2022]
Affiliation(s)
- Berdieke Goemaere
- BCCM/IHEM Fungal Collection Service of Mycology and Aerobiology Sciensano Brussels Belgium
| | - Katrien Lagrou
- Department of Microbiology and Immunology KU Leuven Leuven Belgium
- Clinical Department of Laboratory Medicine National Reference Centre for Mycosis University Hospitals Leuven Leuven Belgium
| | - Isabel Spriet
- Department of Pharmaceutical and Pharmacological Sciences KU Leuven Leuven Belgium
- Pharmacy Department University Hospitals Leuven Leuven Belgium
| | - Marijke Hendrickx
- BCCM/IHEM Fungal Collection Service of Mycology and Aerobiology Sciensano Brussels Belgium
| | - Eline Vandael
- Healthcare‐Associated Infections and Antimicrobial Resistance Sciensano Brussels Belgium
| | - Pierre Becker
- BCCM/IHEM Fungal Collection Service of Mycology and Aerobiology Sciensano Brussels Belgium
| | - Boudewijn Catry
- Healthcare‐Associated Infections and Antimicrobial Resistance Sciensano Brussels Belgium
- Faculty of Medicine Université Libre de Bruxelles (ULB) Brussels Belgium
| |
Collapse
|
26
|
Zhang J, Liu Y, Nie X, Yu Y, Gu J, Zhao L. Trough concentration of itraconazole and its relationship with efficacy and safety: a systematic review and meta-analysis. Infect Drug Resist 2018; 11:1283-1297. [PMID: 30197526 PMCID: PMC6112779 DOI: 10.2147/idr.s170706] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Objectives The optimum trough concentration of itraconazole for clinical response and safty is controversial. The objective of this systematic review and meta-analysis was to determine the optimum trough concentration of itraconazole and evaluate its relationship with efficacy and safety. Methods We searched PubMed, EMBASE, Web of Science, the Cochrane Library, Clinical-Trials.gov, and three Chinese literature databases (CNKI, WanFang, and CBM). We included observational studies that compared clinical outcomes below or above the trough concentration cut-off value which we set as 0.25, 0.5, and 1.0 mg/L. The efficacy outcomes were rate of successful treatment, rate of prophylaxis failure and invasive fungal infection (IFI)-related mortality. The safety outcomes included incidents of hepatotoxicity and other adverse events. Results The study included a total of 29 studies involving 2,346 patients. Our meta-analysis showed that compared with itraconazole trough concentrations (Ctrough) of ≥0.25 mg/L, levels of <0.25 mg/L significantly increased the incidence of IFI for prophylaxis (RR =3.279, 95% confidence interval [CI] 1.73–6.206). Moreover, the success rate of treatment decreased significantly at a cut-off level of 0.5 mg/L (RR =0.396, 95% CI 0.176–0.889). An itraconazole trough level of 1.0 mg/L was associated with hepatotoxicity and other adverse events in a review of many studies. Conclusion An itraconazole trough concentration of 0.25 mg/L should be considered as the lower threshold for prophylaxis, and a target concentration of 0.5 mg/L should be the lower limit for effective treatment. A trough level of 1.0 mg/L is associated with increased hepatotoxicity and other adverse events (using High Performance Liquid Chromatography [HPLC]).
Collapse
Affiliation(s)
- Jingru Zhang
- Clinical Research Center, Beijing Children's Hospital, Capital Medical University, Beijing, China, .,Department of Pharmacy Administration and Clinical Pharmacy, Peking University School of Pharmaceutical Sciences, Beijing, China
| | - Yiwei Liu
- Clinical Research Center, Beijing Children's Hospital, Capital Medical University, Beijing, China,
| | - Xiaolu Nie
- Clinical Research Center, Beijing Children's Hospital, Capital Medical University, Beijing, China,
| | - Yuncui Yu
- Clinical Research Center, Beijing Children's Hospital, Capital Medical University, Beijing, China,
| | - Jian Gu
- Department of Pharmacy, Peking University People's Hospital, Beijing, China
| | - Libo Zhao
- Clinical Research Center, Beijing Children's Hospital, Capital Medical University, Beijing, China,
| |
Collapse
|
27
|
Fluconazole inhibits cellular ergosterol synthesis to confer synergism with berberine against yeast cells. J Glob Antimicrob Resist 2018; 13:125-130. [DOI: 10.1016/j.jgar.2017.12.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 12/06/2017] [Accepted: 12/15/2017] [Indexed: 01/08/2023] Open
|
28
|
Patil S, Majumdar B, Sarode SC, Sarode GS, Awan KH. Oropharyngeal Candidosis in HIV-Infected Patients-An Update. Front Microbiol 2018; 9:980. [PMID: 29867882 PMCID: PMC5962761 DOI: 10.3389/fmicb.2018.00980] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 04/26/2018] [Indexed: 01/16/2023] Open
Abstract
Oropharyngeal candidosis (OPC) is an opportunistic fungal infection that is commonly found in HIV-infected patients, even in the twenty-first century. Candida albicans is the main pathogen, but other Candida species have been isolated. OPC usually presents months or years before other severe opportunistic infections and may indicate the presence or progression of HIV disease. The concept of OPC as a biofilm infection has changed our understanding of its pathobiology. Various anti-fungal agents (both topical and systemic) are available to treat OPC. However, anti-fungal resistance as a result of the long-term use of anti-fungal agents and recurrent oropharyngeal infection in AIDS patients require alternative anti-fungal therapies. In addition, both identifying the causative Candida species and conducting anti-fungal vulnerability testing can improve a clinician's ability to prescribe effective anti-fungal agents. The present review focuses on the current findings and therapeutic challenges for HIV-infected patients with OPC.
Collapse
Affiliation(s)
- Shankargouda Patil
- Division of Oral Pathology, Department of Maxillofacial Surgery and Diagnostic Sciences, College of Dentistry, Jazan University, Jizan, Saudi Arabia
| | - Barnali Majumdar
- Department of Oral Pathology and Microbiology, Bhojia Dental College & Hospital, Baddi, India
| | - Sachin C Sarode
- Department of Oral Pathology and Microbiology, Dr. D.Y. Patil Dental College and Hospital, Dr. D.Y. Patil Vidyapeeth, Pimpri, India
| | - Gargi S Sarode
- Department of Oral Pathology and Microbiology, Dr. D.Y. Patil Dental College and Hospital, Dr. D.Y. Patil Vidyapeeth, Pimpri, India
| | - Kamran H Awan
- College of Dental Medicine, Roseman University of Health Sciences, South Jordan, UT, United States
| |
Collapse
|
29
|
Genetic Diversity and Antifungal Susceptibility of Candida parapsilosis Sensu Stricto Isolated from Bloodstream Infections in Turkish Patients. Mycopathologia 2018; 183:701-708. [PMID: 29725811 DOI: 10.1007/s11046-018-0261-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2017] [Accepted: 04/16/2018] [Indexed: 10/17/2022]
Abstract
Candida parapsilosis sensu stricto is an emerging cause of hospital-acquired Candida infections, predominantly in southern Europe, South America, and Asia. We investigated the genetic diversity and antifungal susceptibility profile of 170 independent C. parapsilosis sensu stricto strains obtained from patients with candidemia who were treated at the Ege University Hospital in Izmir, Turkey, between 2006 and 2014. The identity of each strain was confirmed via PCR amplification and digestion of the secondary alcohol dehydrogenase-encoding gene. The 24-h geometric mean minimum inhibitory concentrations of the antifungal agents, in increasing order, were as follows: posaconazole, 0.10 µg/mL; voriconazole, 0.21 µg/mL; caspofungin, 0.38 µg/mL; amphotericin B, 0.61 µg/mL; anidulafungin, 0.68 µg/mL; and fluconazole, 2.95 µg/mL. Microsatellite genotyping of the isolates (using fluorescently labeled primers and a panel of four different short-nucleotide repeat fragments) identified 25, 17, 17, and 8 different allelic genotypes at the CP6, B5, CP4, and CP1 locus, respectively. Posaconazole, caspofungin, and amphotericin B showed the greatest in vitro activity of the tested systemic azole, echinocandin, and polyene agents, respectively, and the observed antifungal susceptibility of the isolates was shown to be independent of their isolation source. We obtained a combined discriminatory power of 0.99 with a total of 130 genotypes for 170 isolates tested. Finally, microsatellite profiling analysis confirmed the presence of identical genotype between separate isolates, supporting that effective surveillance and infection-prevention programs are essential to limit the impact of C. parapsilosis sensu stricto on hospitalized patients' health.
Collapse
|
30
|
Ullmann AJ, Aguado JM, Arikan-Akdagli S, Denning DW, Groll AH, Lagrou K, Lass-Flörl C, Lewis RE, Munoz P, Verweij PE, Warris A, Ader F, Akova M, Arendrup MC, Barnes RA, Beigelman-Aubry C, Blot S, Bouza E, Brüggemann RJM, Buchheidt D, Cadranel J, Castagnola E, Chakrabarti A, Cuenca-Estrella M, Dimopoulos G, Fortun J, Gangneux JP, Garbino J, Heinz WJ, Herbrecht R, Heussel CP, Kibbler CC, Klimko N, Kullberg BJ, Lange C, Lehrnbecher T, Löffler J, Lortholary O, Maertens J, Marchetti O, Meis JF, Pagano L, Ribaud P, Richardson M, Roilides E, Ruhnke M, Sanguinetti M, Sheppard DC, Sinkó J, Skiada A, Vehreschild MJGT, Viscoli C, Cornely OA. Diagnosis and management of Aspergillus diseases: executive summary of the 2017 ESCMID-ECMM-ERS guideline. Clin Microbiol Infect 2018; 24 Suppl 1:e1-e38. [PMID: 29544767 DOI: 10.1016/j.cmi.2018.01.002] [Citation(s) in RCA: 860] [Impact Index Per Article: 143.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 01/02/2018] [Accepted: 01/03/2018] [Indexed: 02/06/2023]
Abstract
The European Society for Clinical Microbiology and Infectious Diseases, the European Confederation of Medical Mycology and the European Respiratory Society Joint Clinical Guidelines focus on diagnosis and management of aspergillosis. Of the numerous recommendations, a few are summarized here. Chest computed tomography as well as bronchoscopy with bronchoalveolar lavage (BAL) in patients with suspicion of pulmonary invasive aspergillosis (IA) are strongly recommended. For diagnosis, direct microscopy, preferably using optical brighteners, histopathology and culture are strongly recommended. Serum and BAL galactomannan measures are recommended as markers for the diagnosis of IA. PCR should be considered in conjunction with other diagnostic tests. Pathogen identification to species complex level is strongly recommended for all clinically relevant Aspergillus isolates; antifungal susceptibility testing should be performed in patients with invasive disease in regions with resistance found in contemporary surveillance programmes. Isavuconazole and voriconazole are the preferred agents for first-line treatment of pulmonary IA, whereas liposomal amphotericin B is moderately supported. Combinations of antifungals as primary treatment options are not recommended. Therapeutic drug monitoring is strongly recommended for patients receiving posaconazole suspension or any form of voriconazole for IA treatment, and in refractory disease, where a personalized approach considering reversal of predisposing factors, switching drug class and surgical intervention is also strongly recommended. Primary prophylaxis with posaconazole is strongly recommended in patients with acute myelogenous leukaemia or myelodysplastic syndrome receiving induction chemotherapy. Secondary prophylaxis is strongly recommended in high-risk patients. We strongly recommend treatment duration based on clinical improvement, degree of immunosuppression and response on imaging.
Collapse
Affiliation(s)
- A J Ullmann
- Department of Infectious Diseases, Haematology and Oncology, University Hospital Würzburg, Würzburg, Germany; ESCMID Fungal Infection Study Group (EFISG); European Confederation of Medical Mycology (ECMM)
| | - J M Aguado
- Infectious Diseases Unit, University Hospital Madrid, Madrid, Spain; ESCMID Fungal Infection Study Group (EFISG); European Confederation of Medical Mycology (ECMM)
| | - S Arikan-Akdagli
- Department of Medical Microbiology, Hacettepe University Medical School, Ankara, Turkey; ESCMID Fungal Infection Study Group (EFISG); European Confederation of Medical Mycology (ECMM)
| | - D W Denning
- The National Aspergillosis Centre, Wythenshawe Hospital, Mycology Reference Centre Manchester, Manchester University NHS Foundation Trust, ECMM Excellence Centre of Medical Mycology, Manchester, UK; The University of Manchester, Manchester, UK; Manchester Academic Health Science Centre, Manchester, UK; European Confederation of Medical Mycology (ECMM)
| | - A H Groll
- Department of Paediatric Haematology/Oncology, Centre for Bone Marrow Transplantation, University Children's Hospital Münster, Münster, Germany; ESCMID Fungal Infection Study Group (EFISG); European Confederation of Medical Mycology (ECMM)
| | - K Lagrou
- Department of Microbiology and Immunology, ECMM Excellence Centre of Medical Mycology, University Hospital Leuven, Leuven, Belgium; ESCMID Fungal Infection Study Group (EFISG); European Confederation of Medical Mycology (ECMM)
| | - C Lass-Flörl
- Institute of Hygiene, Microbiology and Social Medicine, ECMM Excellence Centre of Medical Mycology, Medical University Innsbruck, Innsbruck, Austria; ESCMID Fungal Infection Study Group (EFISG); European Confederation of Medical Mycology (ECMM)
| | - R E Lewis
- Infectious Diseases Clinic, Sant'Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy; ESCMID Fungal Infection Study Group (EFISG)
| | - P Munoz
- Department of Medical Microbiology and Infectious Diseases, Hospital General Universitario Gregorio Marañón, Madrid, Spain; CIBER Enfermedades Respiratorias - CIBERES (CB06/06/0058), Madrid, Spain; Medicine Department, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain; ESCMID Fungal Infection Study Group (EFISG); European Confederation of Medical Mycology (ECMM)
| | - P E Verweij
- Department of Medical Microbiology, Radboud University Medical Centre, Centre of Expertise in Mycology Radboudumc/CWZ, ECMM Excellence Centre of Medical Mycology, Nijmegen, Netherlands; ESCMID Fungal Infection Study Group (EFISG); European Confederation of Medical Mycology (ECMM)
| | - A Warris
- MRC Centre for Medical Mycology, Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK; ESCMID Fungal Infection Study Group (EFISG); European Confederation of Medical Mycology (ECMM)
| | - F Ader
- Department of Infectious Diseases, Hospices Civils de Lyon, Lyon, France; Inserm 1111, French International Centre for Infectious Diseases Research (CIRI), Université Claude Bernard Lyon 1, Lyon, France; European Respiratory Society (ERS)
| | - M Akova
- Department of Medicine, Section of Infectious Diseases, Hacettepe University Medical School, Ankara, Turkey; ESCMID Fungal Infection Study Group (EFISG); European Confederation of Medical Mycology (ECMM)
| | - M C Arendrup
- Department Microbiological Surveillance and Research, Statens Serum Institute, Copenhagen, Denmark; ESCMID Fungal Infection Study Group (EFISG); European Confederation of Medical Mycology (ECMM)
| | - R A Barnes
- Department of Medical Microbiology and Infectious Diseases, Institute of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK; European Confederation of Medical Mycology (ECMM)
| | - C Beigelman-Aubry
- Department of Diagnostic and Interventional Radiology, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland; European Respiratory Society (ERS)
| | - S Blot
- Department of Internal Medicine, Ghent University, Ghent, Belgium; Burns, Trauma and Critical Care Research Centre, University of Queensland, Brisbane, Australia; European Respiratory Society (ERS)
| | - E Bouza
- Department of Medical Microbiology and Infectious Diseases, Hospital General Universitario Gregorio Marañón, Madrid, Spain; CIBER Enfermedades Respiratorias - CIBERES (CB06/06/0058), Madrid, Spain; Medicine Department, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain; ESCMID Fungal Infection Study Group (EFISG); European Confederation of Medical Mycology (ECMM)
| | - R J M Brüggemann
- Radboud Centre for Infectious Diseases, Radboud University Medical Centre, Centre of Expertise in Mycology Radboudumc/CWZ, ECMM Excellence Centre of Medical Mycology, Nijmegen, Netherlands; ESCMID Fungal Infection Study Group (EFISG)
| | - D Buchheidt
- Medical Clinic III, University Hospital Mannheim, Mannheim, Germany; ESCMID Fungal Infection Study Group (EFISG); European Confederation of Medical Mycology (ECMM)
| | - J Cadranel
- Department of Pneumology, University Hospital of Tenon and Sorbonne, University of Paris, Paris, France; European Respiratory Society (ERS)
| | - E Castagnola
- Infectious Diseases Unit, Istituto Giannina Gaslini Children's Hospital, Genoa, Italy; ESCMID Fungal Infection Study Group (EFISG)
| | - A Chakrabarti
- Department of Medical Microbiology, Postgraduate Institute of Medical Education & Research, Chandigarh, India; European Confederation of Medical Mycology (ECMM)
| | - M Cuenca-Estrella
- Instituto de Salud Carlos III, Madrid, Spain; ESCMID Fungal Infection Study Group (EFISG); European Confederation of Medical Mycology (ECMM)
| | - G Dimopoulos
- Department of Critical Care Medicine, Attikon University Hospital, National and Kapodistrian University of Athens, Medical School, Athens, Greece; European Respiratory Society (ERS)
| | - J Fortun
- Infectious Diseases Service, Ramón y Cajal Hospital, Madrid, Spain; ESCMID Fungal Infection Study Group (EFISG); European Confederation of Medical Mycology (ECMM)
| | - J-P Gangneux
- Univ Rennes, CHU Rennes, Inserm, Irset (Institut de Recherche en santé, environnement et travail) - UMR_S 1085, Rennes, France; ESCMID Fungal Infection Study Group (EFISG); European Confederation of Medical Mycology (ECMM)
| | - J Garbino
- Division of Infectious Diseases, University Hospital of Geneva, Geneva, Switzerland; ESCMID Fungal Infection Study Group (EFISG); European Confederation of Medical Mycology (ECMM)
| | - W J Heinz
- Department of Infectious Diseases, Haematology and Oncology, University Hospital Würzburg, Würzburg, Germany; ESCMID Fungal Infection Study Group (EFISG); European Confederation of Medical Mycology (ECMM)
| | - R Herbrecht
- Department of Haematology and Oncology, University Hospital of Strasbourg, Strasbourg, France; ESCMID Fungal Infection Study Group (EFISG)
| | - C P Heussel
- Diagnostic and Interventional Radiology, Thoracic Clinic, University Hospital Heidelberg, Heidelberg, Germany; European Confederation of Medical Mycology (ECMM)
| | - C C Kibbler
- Centre for Medical Microbiology, University College London, London, UK; European Confederation of Medical Mycology (ECMM)
| | - N Klimko
- Department of Clinical Mycology, Allergy and Immunology, North Western State Medical University, St Petersburg, Russia; European Confederation of Medical Mycology (ECMM)
| | - B J Kullberg
- Radboud Centre for Infectious Diseases, Radboud University Medical Centre, Centre of Expertise in Mycology Radboudumc/CWZ, ECMM Excellence Centre of Medical Mycology, Nijmegen, Netherlands; ESCMID Fungal Infection Study Group (EFISG); European Confederation of Medical Mycology (ECMM)
| | - C Lange
- International Health and Infectious Diseases, University of Lübeck, Lübeck, Germany; Clinical Infectious Diseases, Research Centre Borstel, Leibniz Center for Medicine & Biosciences, Borstel, Germany; German Centre for Infection Research (DZIF), Tuberculosis Unit, Hamburg-Lübeck-Borstel-Riems Site, Lübeck, Germany; European Respiratory Society (ERS)
| | - T Lehrnbecher
- Division of Paediatric Haematology and Oncology, Hospital for Children and Adolescents, Johann Wolfgang Goethe-University, Frankfurt, Germany; European Confederation of Medical Mycology (ECMM)
| | - J Löffler
- Department of Infectious Diseases, Haematology and Oncology, University Hospital Würzburg, Würzburg, Germany; ESCMID Fungal Infection Study Group (EFISG); European Confederation of Medical Mycology (ECMM)
| | - O Lortholary
- Department of Infectious and Tropical Diseases, Children's Hospital, University of Paris, Paris, France; ESCMID Fungal Infection Study Group (EFISG); European Confederation of Medical Mycology (ECMM)
| | - J Maertens
- Department of Haematology, ECMM Excellence Centre of Medical Mycology, University Hospital Leuven, Leuven, Belgium; ESCMID Fungal Infection Study Group (EFISG); European Confederation of Medical Mycology (ECMM)
| | - O Marchetti
- Infectious Diseases Service, Department of Medicine, Lausanne University Hospital, Lausanne, Switzerland; Department of Medicine, Ensemble Hospitalier de la Côte, Morges, Switzerland; ESCMID Fungal Infection Study Group (EFISG); European Confederation of Medical Mycology (ECMM)
| | - J F Meis
- Department of Medical Microbiology and Infectious Diseases, Canisius-Wilhelmina Hospital, Centre of Expertise in Mycology Radboudumc/CWZ, ECMM Excellence Centre of Medical Mycology, Nijmegen, Netherlands; ESCMID Fungal Infection Study Group (EFISG); European Confederation of Medical Mycology (ECMM)
| | - L Pagano
- Department of Haematology, Universita Cattolica del Sacro Cuore, Roma, Italy; European Confederation of Medical Mycology (ECMM)
| | - P Ribaud
- Quality Unit, Pôle Prébloc, Saint-Louis and Lariboisière Hospital Group, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - M Richardson
- The National Aspergillosis Centre, Wythenshawe Hospital, Mycology Reference Centre Manchester, Manchester University NHS Foundation Trust, ECMM Excellence Centre of Medical Mycology, Manchester, UK; The University of Manchester, Manchester, UK; Manchester Academic Health Science Centre, Manchester, UK; ESCMID Fungal Infection Study Group (EFISG); European Confederation of Medical Mycology (ECMM)
| | - E Roilides
- Infectious Diseases Unit, 3rd Department of Paediatrics, Faculty of Medicine, Aristotle University School of Health Sciences, Thessaloniki, Greece; Hippokration General Hospital, Thessaloniki, Greece; ESCMID Fungal Infection Study Group (EFISG); European Confederation of Medical Mycology (ECMM)
| | - M Ruhnke
- Department of Haematology and Oncology, Paracelsus Hospital, Osnabrück, Germany; ESCMID Fungal Infection Study Group (EFISG); European Confederation of Medical Mycology (ECMM)
| | - M Sanguinetti
- Institute of Microbiology, Fondazione Policlinico Universitario A. Gemelli - Università Cattolica del Sacro Cuore, Rome, Italy; ESCMID Fungal Infection Study Group (EFISG); European Confederation of Medical Mycology (ECMM)
| | - D C Sheppard
- Division of Infectious Diseases, Department of Medicine, Microbiology and Immunology, McGill University, Montreal, Canada; ESCMID Fungal Infection Study Group (EFISG); European Confederation of Medical Mycology (ECMM)
| | - J Sinkó
- Department of Haematology and Stem Cell Transplantation, Szent István and Szent László Hospital, Budapest, Hungary; ESCMID Fungal Infection Study Group (EFISG)
| | - A Skiada
- First Department of Medicine, Laiko Hospital, National and Kapodistrian University of Athens, Athens, Greece; ESCMID Fungal Infection Study Group (EFISG); European Confederation of Medical Mycology (ECMM)
| | - M J G T Vehreschild
- Department I of Internal Medicine, ECMM Excellence Centre of Medical Mycology, University Hospital of Cologne, Cologne, Germany; Centre for Integrated Oncology, Cologne-Bonn, University of Cologne, Cologne, Germany; German Centre for Infection Research (DZIF) partner site Bonn-Cologne, Cologne, Germany; European Confederation of Medical Mycology (ECMM)
| | - C Viscoli
- Ospedale Policlinico San Martino and University of Genova (DISSAL), Genova, Italy; ESCMID Fungal Infection Study Group (EFISG); European Confederation of Medical Mycology (ECMM)
| | - O A Cornely
- First Department of Medicine, Laiko Hospital, National and Kapodistrian University of Athens, Athens, Greece; German Centre for Infection Research (DZIF) partner site Bonn-Cologne, Cologne, Germany; CECAD Cluster of Excellence, University of Cologne, Cologne, Germany; Clinical Trials Center Cologne, University Hospital of Cologne, Cologne, Germany; ESCMID Fungal Infection Study Group (EFISG); European Confederation of Medical Mycology (ECMM); ESCMID European Study Group for Infections in Compromised Hosts (ESGICH).
| |
Collapse
|
31
|
Update from a 12-Year Nationwide Fungemia Surveillance: Increasing Intrinsic and Acquired Resistance Causes Concern. J Clin Microbiol 2018; 56:JCM.01564-17. [PMID: 29212705 DOI: 10.1128/jcm.01564-17] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Accepted: 11/30/2017] [Indexed: 12/18/2022] Open
Abstract
New data from the years 2012 to 2015 from the Danish National Fungemia Surveillance are reported, and epidemiological trends are investigated in a 12-year perspective (2004 to 2015). During 2012 to 2015, 1,900 of 1,939 (98%) fungal bloodstream isolates were included. The average incidence was 8.4/100,000 inhabitants, and this appears to represent a stabilizing trend after the increase to 10.1/100,000 in 2011. The incidence was higher in males than females (10.0 versus 6.8) and in patients above 50 years, and those changes were mainly driven by an increasing incidence among 80-to-89-year-old males (65.3/100,000 in 2014 to 2015). The proportion of Candida albicans isolates decreased from 2004 to 2015 (64.4% to 42.4%) in parallel with a doubling of the proportion of Candida glabrata isolates (16.5% to 34.6%, P < 0.0001). C. glabrata was more common among females (34.0% versus 30.4% in males). Following an increase in 2004 to 2011, the annual drug use stabilized during the last 2 to 3 years of that time period but remained higher than in other Nordic countries. This was particularly true for the fluconazole and itraconazole use in the primary health care sector, which exceeded the combined national levels of use of these compounds in each of the other Nordic countries. Fluconazole susceptibility decreased (68.5%, 65.2%, and 60.6% in 2004 to 2007, 2008 to 2011, and 2012 to 2015, respectively, P < 0.0001), and echinocandin resistance emerged in Candida (0%, 0.6%, and 1.7%, respectively, P < 0.001). Amphotericin B susceptibility remained high (98.7%). Among 16 (2.7%) echinocandin-resistant C. glabrata isolates (2012 to 2015), 13 harbored FKS mutations and 5 (31%) were multidrug resistant. The epidemiological changes and the increased incidence of intrinsic and acquired resistance emphasize the importance of continued surveillance and of strengthened focus on antifungal stewardship.
Collapse
|
32
|
Tonin FS, Steimbach LM, Borba HH, Sanches AC, Wiens A, Pontarolo R, Fernandez-Llimos F. Efficacy and safety of amphotericin B formulations: a network meta-analysis and a multicriteria decision analysis. J Pharm Pharmacol 2017; 69:1672-1683. [DOI: 10.1111/jphp.12802] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Accepted: 07/17/2017] [Indexed: 01/25/2023]
Abstract
Abstract
Objectives
Despite its broad spectrum, conventional amphotericin B (AB) is associated with serious adverse events. Lipid-based formulations may offer safer options. We aimed to synthesize the evidence of efficacy and safety of AB formulations.
Methods
We performed a systematic review and network meta-analysis (NMA) to compare all available formulations: conventional AB; lipid complex or ABLC; colloidal dispersion or ABCD; liposomal or LAB; AB in Intralipid. Randomized controlled trials were searched in four databases. Cure, fever, chills, nephrotoxicity, death and drug discontinuation were assessed. NMA was based on Bayesian methods accounting for direct and indirect comparisons. Probability ranks estimating the best formulation were built for each outcome. The relative benefit–risk of formulations was assessed with stochastic multicriteria acceptability analyses (SMAA).
Key findings
We identified 25 trials (n = 2996). No significant differences among drugs were observed for cure or death. All lipid-based formulations were safer than conventional AB for nephrotoxicity. AB-Intralipid was more tolerable than conventional AB and caused less chills than ABCD. AB-Intralipid was the best therapy (>60%) regarding nephrotoxicity, fever, chills and discontinuation. The scenario from SMAA favoured AB-Intralipid (81% acceptability). Conventional AB was secondary to all lipid-based formulations.
Conclusions
Amphotericin B-Intralipid was identified as safer, cost-saving treatment in comparison with other formulations.
Collapse
Affiliation(s)
- Fernanda S Tonin
- Pharmaceutical Sciences Postgraduate Program, Universidade Federal do Paraná, Curitiba, Brazil
| | - Laiza M Steimbach
- Pharmaceutical Sciences Postgraduate Program, Universidade Federal do Paraná, Curitiba, Brazil
| | - Helena H Borba
- Pharmaceutical Sciences Postgraduate Program, Universidade Federal do Paraná, Curitiba, Brazil
| | - Andreia C Sanches
- Department of Pharmacy, Universidade Estadual do Oeste do Paraná, Cascavel, Brazil
| | - Astrid Wiens
- Department of Pharmacy, Universidade Federal do Paraná, Curitiba, Brazil
| | - Roberto Pontarolo
- Department of Pharmacy, Universidade Federal do Paraná, Curitiba, Brazil
| | - Fernando Fernandez-Llimos
- Department of Social Pharmacy, Faculty of Pharmacy, Research Institute for Medicines (iMed.ULisboa), Universidade de Lisboa, Lisbon, Portugal
| |
Collapse
|
33
|
Saunte D, Mrowietz U, Puig L, Zachariae C. Candida
infections in patients with psoriasis and psoriatic arthritis treated with interleukin-17 inhibitors and their practical management. Br J Dermatol 2017; 177:47-62. [DOI: 10.1111/bjd.15015] [Citation(s) in RCA: 175] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/22/2016] [Indexed: 12/13/2022]
Affiliation(s)
- D.M. Saunte
- Department of Dermatology; Zealand University Hospital; Roskilde Denmark
| | - U. Mrowietz
- Psoriasis Center; Department of Dermatology; University Medical Center Schleswig-Holstein; Campus Kiel Germany
| | - L. Puig
- Department of Dermatology; Hospital de la Santa Creu i Sant Pau; Universitat Autònoma de Barcelona; Barcelona Spain
| | - C. Zachariae
- Department of Dermatology and Allergy; Herlev and Gentofte Hospital; University of Copenhagen; Hellerup Denmark
| |
Collapse
|
34
|
Millot M, Girardot M, Dutreix L, Mambu L, Imbert C. Antifungal and Anti-Biofilm Activities of Acetone Lichen Extracts against Candida albicans. Molecules 2017; 22:molecules22040651. [PMID: 28422057 PMCID: PMC6154547 DOI: 10.3390/molecules22040651] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 04/06/2017] [Accepted: 04/14/2017] [Indexed: 01/09/2023] Open
Abstract
Candida albicans is a commensal coloniser of the human gastrointestinal tract and an opportunistic pathogen, especially thanks to its capacity to form biofilms. This lifestyle is frequently involved in infections and increases the yeast resistance to antimicrobials and immune defenses. In this context, 38 lichen acetone extracts have been prepared and evaluated for their activity against C. albicans planktonic and sessile cells. Minimum inhibitory concentrations of extracts (MICs) were determined using the broth microdilution method. Anti-biofilm activity was evaluated using tetrazolium salt (XTT) assay as the ability to inhibit the maturation phase (anti-maturation) or to eradicate a preformed 24 h old biofilm (anti-biofilm). While none of the extracts were active against planktonic cells, biofilm maturation was limited by 11 of the tested extracts. Seven extracts displayed both anti-maturation and anti-biofilm activities (half maximal inhibitory concentrations IC50_mat and IC50_biof ≤ 100 µg/mL); Evernia prunastri and Ramalina fastigiata were the most promising lichens (IC50_mat < 4 µg/mL and IC50_biof < 10 µg/mL). Chemical profiles of the active extracts performed by thin layer chromatography (TLC) and high performance liquid chromatography (HPLC) have been analyzed. Depsides, which were present in large amounts in the most active extracts, could be involved in anti-biofilm activities. This work confirmed that lichens represent a reservoir of compounds with anti-biofilm potential.
Collapse
Affiliation(s)
- Marion Millot
- Laboratoire de Chimie des Substances Naturelles, Faculté de Pharmacie, 2 rue du Dr Marcland, 87025 Limoges, France.
| | - Marion Girardot
- Laboratoire Ecologie et Biologie des Interactions, Equipe Microbiologie de l'Eau, Université de Poitiers, UMR CNRS 7267, F-86073 Poitiers, France.
| | - Lucile Dutreix
- Laboratoire Ecologie et Biologie des Interactions, Equipe Microbiologie de l'Eau, Université de Poitiers, UMR CNRS 7267, F-86073 Poitiers, France.
| | - Lengo Mambu
- Laboratoire de Chimie des Substances Naturelles, Faculté de Pharmacie, 2 rue du Dr Marcland, 87025 Limoges, France.
| | - Christine Imbert
- Laboratoire Ecologie et Biologie des Interactions, Equipe Microbiologie de l'Eau, Université de Poitiers, UMR CNRS 7267, F-86073 Poitiers, France.
| |
Collapse
|
35
|
Poulat C, Nivoix Y, Launoy A, Lutun P, Bachellier P, Rohr S, Woehl ML, Levêque D, Bru V, Herbrecht R, Gourieux B. Assessment of high-priced systemic antifungal prescriptions. Med Mal Infect 2017; 47:382-388. [PMID: 28412043 DOI: 10.1016/j.medmal.2017.03.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Revised: 06/17/2016] [Accepted: 03/10/2017] [Indexed: 11/25/2022]
Abstract
OBJECTIVES To assess compliance with international guidelines for costly antifungal prescriptions and to compare these results with a first study performed in 2007. METHODS Retrospective study including all costly antifungal prescriptions made in surgical and medical intensive care units and in a hepatobiliary, pancreatic, and digestive surgery unit. Prescriptions were assessed in terms of indication, dosage, and antifungal de-escalation. RESULTS Seventy-four treatments were analyzed. Treatments were prescribed for prophylactic (1%), empirical (22%), pre-emptive (16%), or targeted therapy (61%). Caspofungin accounted for 68% of prescriptions, followed by voriconazole (20%) and liposomal amphotericin B (12%). Indication was appropriate in 91%, debatable in 1%, and inappropriate in 8%. Dosage was appropriate in 69%, debatable in 8%, and inappropriate in 23%. Prescriptions were inappropriate for the following reasons: lack of dosage adjustment in light of the hepatic function (10 cases), underdosage or excessive dosage by>25% of the recommended dose in seven cases. De-escalation to fluconazole was implemented in 40% of patients presenting with a fluconazole-susceptible candidiasis. CONCLUSION The overall incidence of appropriate use was higher in 2012 compared with 2007 (62% and 37% respectively, P=0.004). Nevertheless, costly antifungal prescriptions need to be optimized in particular for empirical therapy, dosage adjustment, and potential de-escalation to fluconazole.
Collapse
Affiliation(s)
- C Poulat
- Pharmacy, hôpitaux universitaires de Strasbourg, université de Strasbourg, 67098 Strasbourg, France
| | - Y Nivoix
- Pharmacy, hôpitaux universitaires de Strasbourg, université de Strasbourg, 67098 Strasbourg, France.
| | - A Launoy
- Surgical Intensive Care Department, hôpitaux universitaires de Strasbourg, université de Strasbourg, 67098 Strasbourg, France
| | - P Lutun
- Medical Intensive Care Department, hôpitaux universitaires de Strasbourg, université de Strasbourg, 67098 Strasbourg, France
| | - P Bachellier
- Hepatobiliary, Pancreatic and Digestive Surgery Department, hôpitaux universitaires de Strasbourg, université de Strasbourg, 67098 Strasbourg, France
| | - S Rohr
- Hepatobiliary, Pancreatic and Digestive Surgery Department, hôpitaux universitaires de Strasbourg, université de Strasbourg, 67098 Strasbourg, France
| | - M-L Woehl
- Hepatobiliary, Pancreatic and Digestive Surgery Department, hôpitaux universitaires de Strasbourg, université de Strasbourg, 67098 Strasbourg, France
| | - D Levêque
- Pharmacy, hôpitaux universitaires de Strasbourg, université de Strasbourg, 67098 Strasbourg, France
| | - V Bru
- Institut de parasitologie et de pathologie tropicale, hôpitaux universitaires de Strasbourg, université de Strasbourg, 67098 Strasbourg, France
| | - R Herbrecht
- Oncology and Hematology Department, hôpitaux universitaires de Strasbourg, université de Strasbourg, 67098 Strasbourg, France
| | - B Gourieux
- Pharmacy, hôpitaux universitaires de Strasbourg, université de Strasbourg, 67098 Strasbourg, France
| |
Collapse
|
36
|
Chang CC, Slavin MA, Chen SCA. New developments and directions in the clinical application of the echinocandins. Arch Toxicol 2017; 91:1613-1621. [DOI: 10.1007/s00204-016-1916-3] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 12/13/2016] [Indexed: 01/05/2023]
|
37
|
Shao B, Ma Y, Li Q, Wang Y, Zhu Z, Zhao H, Sun J, Dong L, Zhu Y, Zhao N, Qin Y. Effects of cytochrome P450 3A4 and non-genetic factors on initial voriconazole serum trough concentrations in hematological patients with different cytochrome P450 2C19 genotypes. Xenobiotica 2017; 47:1121-1129. [PMID: 27937048 DOI: 10.1080/00498254.2016.1271960] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
1. Polymorphisms of cytochrome P450 2C19 (CYP2C19) is an important factor contributing to variability of voriconazole pharmacokinetics. Polymorphisms of CYP3A4, CYP3A5, CYP2C9 and non-genetic factors such as age, gender, body mass index (BMI), transaminase levels, concomitant medications might also affect voriconazole initial steady serum trough concentration (VICmin) in haematological patients, but the effects were not clear. 2. Eighteen single-nucleotide polymorphisms in CYP2C19, CYP3A4, CYP3A5, CYP2C9 were genotyped. Patients were stratified into two groups according to CYP2C19 genotype. Group 1 were patients with CYP2C19*2 or CYP2C19*3, and Group 2 were homozygous extensive metabolizers. The effects were studied in different groups. VICmin was adjusted on daily dose (VICmin/D) for overcoming effect of dose. 3. A total of 106 blood samples from 86 patients were included. In final optimal scaling regression models, polymorphisms of rs4646437 (CYP3A4), age, BMI was identified to be factors of VICmin/D in Group 1 (R2 = .255, p < .001). Only age was confirmed as a factor of VICmin/D in Group 2 (R2 = 0.144, p = .021). 4. Besides polymorphisms of CYP2C19, in individualized medication of voriconazole in haematological patients, polymorphisms of CYP3A4, and non-genetic factors as BMI, age should also be taken into account, especially for individuals with CYP2C19*2 or CYP2C19*3.
Collapse
Affiliation(s)
| | | | | | | | - Zunmin Zhu
- b Department of Haematology , Zhengzhou University People's Hospital , Zhengzhou , Henan , PR China
| | | | - Jun Sun
- a Department of Pharmacy and
| | | | | | | | | |
Collapse
|
38
|
|
39
|
De Wit S, Clumeck N. Opportunistic Infections. Infect Dis (Lond) 2017. [DOI: 10.1016/b978-0-7020-6285-8.00094-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|
40
|
Bandara HMHN, Matsubara VH, Samaranayake LP. Future therapies targeted towards eliminating Candida biofilms and associated infections. Expert Rev Anti Infect Ther 2016; 15:299-318. [PMID: 27927053 DOI: 10.1080/14787210.2017.1268530] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Candida species are common human commensals and cause either superficial or invasive opportunistic infections. The biofilm form of candida as opposed to its suspended, planktonic form, is predominantly associated with these infections. Alternative or adjunctive therapies are urgently needed to manage Candida infections as the currently available short arsenal of antifungal drugs has been compromised due to their systemic toxicity, cross-reactivity with other drugs, and above all, by the emergence of drug-resistant Candida species due to irrational drug use. Areas covered: Combination anti-Candida therapies, antifungal lock therapy, denture cleansers, and mouth rinses have all been proposed as alternatives for disrupting candidal biofilms on different substrates. Other suggested approaches for the management of candidiasis include the use of natural compounds, such as probiotics, plants extracts and oils, antifungal quorum sensing molecules, anti-Candida antibodies and vaccines, cytokine therapy, transfer of primed immune cells, photodynamic therapy, and nanoparticles. Expert commentary: The sparsity of currently available antifungals and the plethora of proposed anti-candidal therapies is a distinct indication of the urgent necessity to develop efficacious therapies for candidal infections. Alternative drug delivery approaches, such as probiotics, reviewed here is likely to be a reality in clinical settings in the not too distant future.
Collapse
Affiliation(s)
- H M H N Bandara
- a School of Dentistry , The University of Queensland , Herston , QLD , Australia
| | - V H Matsubara
- b School of Dentistry , University of São Paulo , São Paulo , SP , Brazil.,c Department of Microbiology, Institute of Biomedical Sciences , University of São Paulo , São Paulo , SP , Brazil
| | - L P Samaranayake
- a School of Dentistry , The University of Queensland , Herston , QLD , Australia.,d Faculty of Dentistry , University of Kuwait , Kuwait
| |
Collapse
|
41
|
Wiederhold NP. Echinocandin Resistance in Candida Species: a Review of Recent Developments. Curr Infect Dis Rep 2016; 18:42. [PMID: 27771864 DOI: 10.1007/s11908-016-0549-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The echinocandins are important agents for the treatment of invasive fungal infections, especially those caused by Candida species. However, as with other antimicrobial agents, microbiologic resistance to this class of antifungal agents has emerged and can result in clinical failure. Several studies have recently reported an increase in echinocandin resistance in Candida glabrata isolates at various medical centers in different geographic regions of the USA. Recent studies have also reported that many of these isolates may also be fluconazole resistant, leaving few treatment options available for clinicians to use in patients with invasive candidiasis caused by this species. Our understanding of the clinical relevance of specific point mutations within the FKS genes that cause echinocandin resistance and risk factors for the development of microbiologic resistance and clinical failure have also increased. The purpose of this review is to discuss echinocandin resistance in Candida species and recent reports that have increased our understanding of this growing clinical problem.
Collapse
Affiliation(s)
- Nathan P Wiederhold
- Department of Pathology, Fungus Testing Laboratory, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX, 78229, USA.
| |
Collapse
|
42
|
Minea B, Nastasa V, Kolecka A, Mares M, Marangoci N, Rosca I, Pinteala M, Hancianu M, Mares M. Etiologic Agents and Antifungal Susceptibility of Oral Candidosis from Romanian patients with HIV-infection or type 1 diabetes mellitus. Pol J Microbiol 2016; 65:123-9. [PMID: 27282005 DOI: 10.5604/17331331.1197327] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
This is the first Romanian investigation of oral candidosis in patients suffering of HIV-infection or type 1 diabetes mellitus (T1DM). Candida albicans was the dominant species in both types of isolates: n = 14 (46.7%) in T1DM, n = 60 (69.8%) in HIV. The most frequent non-albicans Candida spp. were Candida kefyr (n = 6; 20%) in T1DM and Candida dubliniensis (n = 8; 9.3%) in HIV. Resistance to fluconazole was detected only in the HIV non-albicans Candida group (n = 8; 9.3%). All isolates were susceptible to VOR. The experimental drug MXP had MIC values equal or close to the ones of VOR. Echinocandin resistance was more frequent than azole resistance.
Collapse
|
43
|
Abstract
In humans, microbial cells (including bacteria, archaea, and fungi) greatly outnumber host cells. Candida albicans is the most prevalent fungal species of the human microbiota; this species asymptomatically colonizes many areas of the body, particularly the gastrointestinal and genitourinary tracts of healthy individuals. Alterations in host immunity, stress, resident microbiota, and other factors can lead to C. albicans overgrowth, causing a wide range of infections, from superficial mucosal to hematogenously disseminated candidiasis. To date, most studies of C. albicans have been carried out in suspension cultures; however, the medical impact of C. albicans (like that of many other microorganisms) depends on its ability to thrive as a biofilm, a closely packed community of cells. Biofilms are notorious for forming on implanted medical devices, including catheters, pacemakers, dentures, and prosthetic joints, which provide a surface and sanctuary for biofilm growth. C. albicans biofilms are intrinsically resistant to conventional antifungal therapeutics, the host immune system, and other environmental perturbations, making biofilm-based infections a significant clinical challenge. Here, we review our current knowledge of biofilms formed by C. albicans and closely related fungal species.
Collapse
Affiliation(s)
- Clarissa J Nobile
- Department of Molecular and Cell Biology, School of Natural Sciences, University of California, Merced, California 95343;
| | - Alexander D Johnson
- Department of Microbiology and Immunology, University of California, San Francisco, California 94143;
| |
Collapse
|
44
|
Jin H, Wang T, Falcione BA, Olsen KM, Chen K, Tang H, Hui J, Zhai S. Trough concentration of voriconazole and its relationship with efficacy and safety: a systematic review and meta-analysis. J Antimicrob Chemother 2016; 71:1772-85. [PMID: 26968880 PMCID: PMC4896404 DOI: 10.1093/jac/dkw045] [Citation(s) in RCA: 116] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Accepted: 01/29/2016] [Indexed: 01/12/2023] Open
Abstract
This meta-analysis showed trough concentrations of 0.5 mg/L to be the lower limit of voriconazole during treatment, whereas trough concentrations of 3.0 mg/L were associated with an increased risk of moderate to severe hepatotoxicity, particularly for the Asian population. Objectives The optimum trough concentration of voriconazole for clinical response and safety is controversial. The objective of this review was to determine the optimum trough concentration of voriconazole and evaluate its relationship with efficacy and safety. Methods MEDLINE, EMBASE, ClinicalTrials.gov, the Cochrane Library and three Chinese literature databases were searched. Observational studies that compared clinical outcomes below and above the trough concentration cut-off value were included. We set the trough concentration cut-off value for efficacy as 0.5, 1.0, 1.5, 2.0 and 3.0 mg/L and for safety as 3.0, 4.0, 5.0, 5.5 and 6.0 mg/L. The efficacy outcomes were invasive fungal infection-related mortality, all-cause mortality, rate of successful treatment and rate of prophylaxis failure. The safety outcomes included incidents of hepatotoxicity, neurotoxicity and visual disorders. Results A total of 21 studies involving 1158 patients were included. Compared with voriconazole trough concentrations of >0.5 mg/L, levels of <0.5 mg/L significantly decreased the rate of treatment success (risk ratio = 0.46, 95% CI 0.29–0.74). The incidence of hepatotoxicity was significantly increased with trough concentrations >3.0, >4.0, >5.5 and >6.0 mg/L. The incidence of neurotoxicity was significantly increased with trough concentrations >4.0 and >5.5 mg/L. Conclusions A voriconazole level of 0.5 mg/L should be considered the lower threshold associated with efficacy. A trough concentration >3.0 mg/L is associated with increased hepatotoxicity, particularly for the Asian population, and >4.0 mg/L is associated with increased neurotoxicity.
Collapse
Affiliation(s)
- Haiying Jin
- Department of Pharmacy, Peking University Third Hospital, Beijing, China Department of Pharmacy, The Affiliated Hospital of Medical College, Ningbo University, Ningbo, Zhejiang, China
| | - Tiansheng Wang
- Department of Pharmacy, Peking University Third Hospital, Beijing, China Department of Pharmacy Administration and Clinical Pharmacy, Peking University School of Pharmaceutical Sciences, Beijing, China
| | - Bonnie A Falcione
- Department of Pharmacy and Therapeutics, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, USA
| | - Keith M Olsen
- Department of Pharmacy Practice, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Ken Chen
- Department of Pharmacy, Peking University Third Hospital, Beijing, China Department of Pharmacy Administration and Clinical Pharmacy, Peking University School of Pharmaceutical Sciences, Beijing, China
| | - Huilin Tang
- Department of Pharmacy, Peking University Third Hospital, Beijing, China
| | - John Hui
- Department of Pharmacy, Stanford University Hospital and Clinics, Palo Alto, CA, USA
| | - Suodi Zhai
- Department of Pharmacy, Peking University Third Hospital, Beijing, China
| |
Collapse
|
45
|
Onishi A, Iwasaku M, Sato A, Furukawa TA. Interventions for the management of esophageal candidiasis in immunocompromised patients. Hippokratia 2015. [DOI: 10.1002/14651858.cd011938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
- Akira Onishi
- Kyoto University Graduate School of Medicine / School of Public Health; Department of Health Promotion and Human Behavior; Yoshida Konoe-cho, Sakyo-ku Kyoto Japan 606-8501
| | - Masahiro Iwasaku
- Kyoto University Graduate School of Medicine / School of Public Health; Department of Pharmacoepidemiology; Yoshida Konoe-cho, Sakyo-ku Kyoto Japan 606-8501
| | - Akira Sato
- Kyoto University Graduate School of Medicine / School of Public Health; Department of Pharmacoepidemiology; Yoshida Konoe-cho, Sakyo-ku Kyoto Japan 606-8501
| | - Toshi A Furukawa
- Kyoto University Graduate School of Medicine / School of Public Health; Department of Health Promotion and Human Behavior; Yoshida Konoe-cho, Sakyo-ku Kyoto Japan 606-8501
| |
Collapse
|
46
|
Astvad K, Johansen HK, Høiby N, Steptoe P, Ishøy T. Oropharyngeal Candidiasis in Palliative Care Patients in Denmark. J Palliat Med 2015; 18:940-4. [DOI: 10.1089/jpm.2015.29003.ka] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Karen Astvad
- Unit of Mycology, Statens Serum Institut, Copenhagen, Denmark
- Department of Clinical Microbiology, Rigshospitalet, Copenhagen, Denmark
| | | | - Niels Høiby
- Department of Clinical Microbiology, Rigshospitalet, Copenhagen, Denmark
| | | | | |
Collapse
|
47
|
Grau S, Pozo JC, Romá E, Salavert M, Barrueta JA, Peral C, Rodriguez I, Rubio-Rodríguez D, Rubio-Terrés C. Cost-effectiveness of three echinocandins and fluconazole in the treatment of candidemia and/or invasive candidiasis in nonneutropenic adult patients. CLINICOECONOMICS AND OUTCOMES RESEARCH 2015; 7:527-35. [PMID: 26508881 PMCID: PMC4610791 DOI: 10.2147/ceor.s91587] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
OBJECTIVE To estimate the cost-effectiveness of three echinocandins (anidulafungin, caspofungin, and micafungin) and generic fluconazole in the treatment of nonneutropenic adult patients with candidemia and/or invasive candidiasis in intensive care units in Spain. MATERIALS AND METHODS A decision-tree model was applied. The success and safety (hepatic and renal adverse effects) of first-line treatments were obtained from meta-analyses and systematic reviews of clinical trials. In the case of failure, a second-line treatment (liposomal amphotericin B after the echinocandins, or one of the echinocandins after fluconazole) was administered. The duration of the treatments (14 days total) was established by a panel of clinical experts using the Delphi method and according to Infectious Diseases Society of America guidelines. The cost of the medications and renal toxicity were considered. Deterministic and probabilistic sensitivity analysis using Monte Carlo simulations were carried out. RESULTS The total cost of the treatment of candidemia and/or invasive candidiasis with anidulafungin, caspofungin, micafungin, and fluconazole was €5,483, €5,968, €6,231, and €2,088, respectively. Anidulafungin was the dominant treatment (more effective, less expensive) compared to micafungin and caspofungin. The cost of achieving one more patient successfully treated with anidulafungin, caspofungin, and micafungin compared to fluconazole was €17,199, €23,962, and €27,339, respectively. The result remained stable, despite modification of the duration of the first-line and second-line treatments, as well as most of the dosing regimens. The probabilistic analysis also remained stable. CONCLUSION In accordance with this economic study, anidulafungin would produce savings and would be the dominant treatment compared with micafungin and caspofungin in nonneutropenic adult patients with candidemia and/or invasive candidiasis in intensive care units in Spain.
Collapse
Affiliation(s)
- S Grau
- Hospital del Mar (IMIM), Barcelona, Spain
| | - JC Pozo
- Hospital Universitario Reina Sofía, Córdoba, Spain
| | - E Romá
- Hospital Universitario Reina Sofía, Córdoba, Spain
| | - M Salavert
- Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | | | - C Peral
- Pfizer SLU, Alcobendas, Spain
| | | | | | | |
Collapse
|
48
|
Current strategies for prevention of oral manifestations of human immunodeficiency virus. Oral Surg Oral Med Oral Pathol Oral Radiol 2015; 121:29-38. [PMID: 26679357 DOI: 10.1016/j.oooo.2015.09.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Revised: 07/08/2015] [Accepted: 09/02/2015] [Indexed: 01/03/2023]
Abstract
OBJECTIVES Strategies to prevent new-onset and recurrent oral manifestations of human immunodeficiency virus (HIV), including fungal, viral, neoplastic, and idiopathic mucosal diseases and destructive periodontal conditions, are poorly understood. STUDY DESIGN A structured review of the English language literature in PubMed through March 2015 was conducted to identify current prevention strategies for initial and recurrent oral manifestations of HIV. RESULTS Pharmacologic approaches, including combination antiretroviral therapy or other targeted therapies for prevention of oropharyngeal candidiasis, orolabial herpes, oral hairy leukoplakia, oral Kaposi sarcoma, linear gingival erythema and necrotizing ulcerative periodontitis were found. Nonpharmacologic approaches for prevention of oropharyngeal candidiasis, orolabial herpes, oral hairy leukoplakia, and necrotizing ulcerative periodontitis are presented. CONCLUSIONS Current strategies for the prevention of oral manifestations of HIV include pharmacologic and nonpharmacologic therapies. Immune reconstitution inflammatory syndrome, future vaccine therapy for pathogens causing oral mucosal disease, and the possible role of oral inflammatory disease prevention in controlling HIV disease progression are discussed.
Collapse
|
49
|
Abstract
PURPOSE OF REVIEW Chronic infections of the small intestine cause significant morbidity and mortality globally. This review focuses on the recent advances in the field of our understanding of selected intestinal infections. RECENT FINDINGS Primary and secondary immunodeficiency increase the susceptibility to many chronic intestinal infections. Endoscopy and intestinal biopsies are central to establishing a diagnosis of these conditions. Tuberculosis (TB) remains a major global health challenge. Emerging therapeutic agents to counteract multidrug-resistant strains have shown clinical efficacy, but concerns regarding mortality remain. PCR-based diagnostic TB tests have the potential to reduce diagnostic delays, but remain to be validated for intestinal infections. Adjunctive diagnostic imaging modalities can differentiate infections from Crohn's disease with increasing accuracy. Whipple's disease remains rare, but there have been substantial advances in our understanding of the causative organism Tropheryma whipplei. Extended treatment with broad-spectrum antibiotics is effective in most cases. The narrow therapeutic window and limited armamentarium for treating invasive filamentous fungal infections contribute to their significant morbidity and high rates of mortality. SUMMARY The speed and accuracy of diagnosing chronic intestinal infections have improved with recent imaging and laboratory methodologies. Significant research opportunities remain for clinicians and scientists to improve the diagnostic accuracy and clinical outcomes of chronic intestinal infections.
Collapse
Affiliation(s)
- Billy Bourke
- aNational Centre for Paediatric Gastroenterology, Our Lady's Children's Hospital bNational Children's Research Centre, Crumlin, Dublin cUCD School of Medicine and Medical Science dConway Institute, University College Dublin, Belfield, Dublin, Ireland
| | | |
Collapse
|
50
|
Hospital use of systemic antifungal drugs: a multi-center surveillance update from Germany. Infection 2015; 43:423-9. [PMID: 25687588 DOI: 10.1007/s15010-015-0742-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Accepted: 02/02/2015] [Indexed: 10/24/2022]
Abstract
BACKGROUND The consumption of antifungal agents increased over the last decade, resulting in the development of resistant organisms and causing a significant pharmaco economic burden. Antifungal drugs are widely used for the treatment of systemic fungal infections and high-risk patients, especially with severe hematological or oncological conditions. Up to date, there are no reliable and systematically reported data on the consumption of antifungal substances on a nationwide level available. The presented study gives an update to the previously published multicenter study investigating antifungal consumption in different settings from five university hospital centers in Germany from 2001 to 2003. METHODS Consumption data for systemic antifungal drugs were obtained through the hospital pharmacies for 2001-2003 and 2008-2011 regarding the medical and surgical services of five university hospital centers in Germany (A-E). Drug use densities were calculated as yearly RDDs/100 patient days. These calculations were performed for the surgical and medical services, and independently for surgical and medical ICUs, as well as for the hematology-oncology services. RESULTS We report an increased utilization of systemic antifungal drugs in both study periods. The mean drug use density (mean value of all 5 hospitals) in the medical services increased by 24% between 2001 and 2003. In 2011, this value was 37% above the level from 2001 (12.4 RDD/100 patient days in 2001, 15.4 RDD/100 patient days in 2003, 17.0 RDD/100 patient days in 2011). The 4-year average drug use density (2008-2011) of medical services ranged between 11.6 RDD/100 patient days (hospital E) and 23.8 RDD/100 patient days (hospital A). Drug use densities were in medical intensive care units 29.4 RDD/100 patient days and hematology-oncology services 49.9 RDD/100 patient days. CONCLUSIONS Despite the variability of the prescribing patterns between the tertiary hospitals, the presented pharmaco-epidemiological data are a cornerstone for the initiation and implementation of effective antifungal stewardship programmes and might serve as important benchmarking information for other hospitals with similar structures and baseline settings.
Collapse
|