1
|
Nussbaumer-Pröll A, Matzneller P, Eberl S, Zeitlinger M. Pulmonary surfactant impacts in vitro activity of selected antifungal drugs against Candida krusei and Candida albicans. Eur J Clin Microbiol Infect Dis 2024; 43:927-936. [PMID: 38483681 PMCID: PMC11109016 DOI: 10.1007/s10096-024-04799-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 03/05/2024] [Indexed: 05/22/2024]
Abstract
PURPOSE This study investigates how surfactants affect the in-vitro anti-infective efficacy of micafungin, caspofungin, anidulafungin, and amphotericin B in treating pulmonary mycoses. METHODS MIC values for antifungal agents were determined against Candida krusei (now Pichia kudriavzevii) ATCC 6258, Candida albicans ATCC 90028, and 18 clinical isolates using the broth microdilution method in RPMI medium, following EUCAST recommendations. MIC assays included testing with and without Curosurf® surfactant at 1 mg/mL for C. krusei ATCC 6258 and all C. krusei isolates. Subsequent Time-kill studies in Sabouraud broth involved testing both C. albicans ATCC 90028 and C. krusei ATCC 6258 strains at concentrations equal their respective MIC values, with and without surfactant, using all four antifungals. CFU/mL were assessed at multiple time points up to 24 h. TKCs with different surfactant concentrations for C. krusei ATCC 6258 and mini-TKCs at various concentrations relative to the MIC of C. krusei isolates and the reference strain were conducted with micafungin, anidulafungin, and caspofungin. RESULTS MIC results showed that 1 µg/mL surfactant reduced killing of micafungin and anidulafungin against C. krusei, while caspofungin was unaffected. Amphotericin B's MIC decreased by half. TKCs demonstrated significant effects of surfactant on micafungin and anidulafungin against C. krusei, with complete abolition of anidulafungin's activity against C. albicans. CONCLUSION This in-vitro study highlights the concentration-dependent inhibitory effect of surfactant on antifungal activity against C. krusei and, to some extent, C. albicans, necessitating further clinical validation for invasive lung mycoses treatment.
Collapse
Affiliation(s)
- Alina Nussbaumer-Pröll
- Department of Clinical Pharmacology, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
| | - Peter Matzneller
- Department of Clinical Pharmacology, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
| | - Sabine Eberl
- Department of Clinical Pharmacology, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
| | - Markus Zeitlinger
- Department of Clinical Pharmacology, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria.
| |
Collapse
|
2
|
Hoenigl M, Lewis R, van de Veerdonk FL, Verweij PE, Cornely OA. Liposomal amphotericin B—the future. J Antimicrob Chemother 2022; 77:ii21-ii34. [PMID: 36426674 PMCID: PMC9693803 DOI: 10.1093/jac/dkac353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 09/26/2022] [Indexed: 11/26/2022] Open
Abstract
Advances in medicine have led to a growing number of people with compromised or suppressed immune systems who are susceptible to invasive fungal infections. In particular, severe fungal infections are becoming increasingly common in ICUs, affecting people within and outside of traditional risk groups alike. This is exemplified by the emergence of severe viral pneumonia as a significant risk factor for invasive pulmonary aspergillosis, and the recognition of influenza-associated pulmonary aspergillosis and, more recently, COVID-19-associated pulmonary aspergillosis. The treatment landscape for haematological malignancies has changed considerably in recent years, and some recently introduced targeted agents, such as ibrutinib, are increasing the risk of invasive fungal infections. Consideration must also be given to the risk of drug–drug interactions between mould-active azoles and small-molecule kinase inhibitors. At the same time, infections caused by rare moulds and yeasts are increasing, and diagnosis continues to be challenging. There is growing concern about azole resistance among both moulds and yeasts, mandating continuous surveillance and personalized treatment strategies. It is anticipated that the epidemiology of fungal infections will continue to change and that new populations will be at risk. Early diagnosis and appropriate treatment remain the most important predictors of survival, and broad-spectrum antifungal agents will become increasingly important. Liposomal amphotericin B will remain an essential therapeutic agent in the armamentarium needed to manage future challenges, given its broad antifungal spectrum, low level of acquired resistance and limited potential for drug–drug interactions.
Collapse
Affiliation(s)
- M Hoenigl
- Division of Infectious Diseases, Department of Internal Medicine, Medical University of Graz , Graz , Austria
- BioTechMed-Graz , Graz , Austria
- European Confederation of Medical Mycology (ECMM) Excellence Center, Medical University of Graz , Graz , Austria
| | - R Lewis
- Department of Medical and Surgical Sciences, Infectious Diseases Hospital, IRCSS S’Orsola-Malpighi, University of Bologna , Bologna , Italy
| | - F L van de Veerdonk
- Department of Internal Medicine, Radboud Center for Infectious Diseases, Radboud University Medical Center , Nijmegen , The Netherlands
| | - P E Verweij
- Department of Medical Microbiology, Radboud University Medical Center—CWZ Center of Expertise for Mycology , Nijmegen , The Netherlands
- Center for Infectious Disease Research, Diagnostics and Laboratory Surveillance, National Institute for Public Health and the Environment (RIVM) , Bilthoven , The Netherlands
| | - O A Cornely
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Translational Research, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) , Cologne , Germany
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD) and Excellence Center for Medical Mycology (ECMM) , Cologne , Germany
- German Centre for Infection Research (DZIF), Partner Site Bonn-Cologne , Cologne , Germany
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Clinical Trials Centre Cologne (ZKS Köln) , Cologne , Germany
| |
Collapse
|
3
|
Xie Y, Zhou X, Zhang J, Yu H, Song Z. Immunomodulatory responses of differentially polarized macrophages to fungal infections. Int Immunopharmacol 2022; 111:109089. [PMID: 35964406 DOI: 10.1016/j.intimp.2022.109089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 07/16/2022] [Accepted: 07/22/2022] [Indexed: 11/05/2022]
Abstract
Macrophages, the first line of defense against invasive fungi in the innate immune system, are widely distributed in the blood and tissues of the body. In response to various internal and external stimulators, macrophages can polarize into classically activated macrophages (M1) and alternatively activated macrophages (M2). These two types of polarized macrophages play different roles in antifungal activity and in maintaining the steady-state balance between inflammation and tissue repair. However, the antifungal mechanisms of M1- and M2-type macrophages have not been fully described. In this review, the immune regulatory mechanisms against pathogenic fungi of these two classical types of macrophages in various tissues are summarized. The effects of antifungal factors on macrophage differentiation are also highlighted. The description of these data, on the one hand provides valuable insight for future investigations and also highlights new strategies for the treatment of pathogenic fungal infections.
Collapse
Affiliation(s)
- Yuxin Xie
- School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, PR China.
| | - Xue Zhou
- School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, PR China.
| | - Jinping Zhang
- School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, PR China; The Public Platform of Molecular Biotechnology, Public Center of Experimental Technology, Southwest Medical University, Luzhou 646000, People's Republic of China.
| | - Hong Yu
- School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, PR China; The Public Platform of Cell Biotechnology, Public Center of Experimental Technology, Southwest Medical University, Luzhou 646000, PR China.
| | - Zhangyong Song
- School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, PR China; The Public Platform of Molecular Biotechnology, Public Center of Experimental Technology, Southwest Medical University, Luzhou 646000, People's Republic of China.
| |
Collapse
|
4
|
Rauwolf KK, Hoertnagl C, Lass-Floerl C, Groll AH. Interaction in vitro of pulmonary surfactant with antifungal agents used for treatment and prevention of invasive aspergillosis. J Antimicrob Chemother 2021; 77:695-698. [PMID: 34788449 DOI: 10.1093/jac/dkab422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 10/19/2021] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Optimizing antifungal therapy is important to improve outcomes in severely immunocompromised patients. OBJECTIVES We analysed the in vitro interaction between pulmonary surfactant and antifungal agents used for management of invasive pulmonary aspergillosis. METHODS Amphotericin B formulations, mould-active triazoles and echinocandins were tested in vitro against 24 clinical isolates of different Aspergillus spp. with and without the addition of a commercial porcine surfactant (Curosurf®; Poractant alfa, Nycomed, Austria). The data are presented as MIC or minimum effective concentration (MEC) ranges, as MIC or MEC values that inhibited 90% of the isolates (MIC90 or MEC90) and as geometric mean (GM) MIC or MEC values. RESULTS For amphotericin B products, addition of surfactant to a final concentration of 10% led to a statistically significant reduction of the GM MIC for all Aspergillus isolates tested after 24 h (0.765 versus 0.552 mg/L; P < 0.05). For the mould-active triazoles, addition of 10% surfactant resulted in a significantly higher GM MIC at 48 h (0.625 versus 0.898 mg/L; P < 0.05). For the echinocandins, the addition of 10% surfactant led to a significantly higher GM MEC after both 24 h (0.409 versus 0.6532 mg/L; P < 0.01) and 48 h (0.527 versus 0.9378 mg/L; P < 0.01). There were no meaningful differences between individual members of the three existing classes of antifungal agents or between the different Aspergillus spp. tested. CONCLUSIONS Using EUCAST methodology, addition of porcine surfactant up to a concentration of 10% had a minor, and presumably non-relevant, impact on the in vitro activity of antifungal agents used in prophylaxis and treatment of invasive pulmonary aspergillosis.
Collapse
Affiliation(s)
- Kerstin K Rauwolf
- Infectious Disease Research Program, Center for Bone Marrow Transplantation and Department of Paediatric Haematology/Oncology, University Children's Hospital Münster, Münster, Germany.,Division of Pediatric Oncology, University Children's Hospital, Zürich, Switzerland
| | - Caroline Hoertnagl
- Institute of Hygiene and Medical Microbiology, Christian Doppler Laboratory for Invasive Fungal Infections, Medical University of Innsbruck, Innsbruck, Austria
| | - Cornelia Lass-Floerl
- Institute of Hygiene and Medical Microbiology, Christian Doppler Laboratory for Invasive Fungal Infections, Medical University of Innsbruck, Innsbruck, Austria
| | - Andreas H Groll
- Infectious Disease Research Program, Center for Bone Marrow Transplantation and Department of Paediatric Haematology/Oncology, University Children's Hospital Münster, Münster, Germany
| |
Collapse
|
5
|
Adler-Moore J, Lewis RE, Brüggemann RJM, Rijnders BJA, Groll AH, Walsh TJ. Preclinical Safety, Tolerability, Pharmacokinetics, Pharmacodynamics, and Antifungal Activity of Liposomal Amphotericin B. Clin Infect Dis 2020; 68:S244-S259. [PMID: 31222254 PMCID: PMC6495008 DOI: 10.1093/cid/ciz064] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The improved safety profile and antifungal efficacy of liposomal amphotericin B (LAmB) compared to conventional amphotericin B deoxycholate (DAmB) is due to several factors including, its chemical composition, rigorous manufacturing standards, and ability to target and transit through the fungal cell wall. Numerous preclinical studies have shown that LAmB administered intravenously distributes to tissues frequently infected by fungi at levels above the minimum inhibitory concentration (MIC) for many fungi. These concentrations can be maintained from one day to a few weeks, depending upon the tissue. Tissue accumulation is dose-dependent with drug clearance occurring most rapidly from the brain and slowest from the liver and spleen. LAmB localizes in lung epithelial lining fluid, within liver and splenic macrophages and in kidney distal tubules. LAmB has been used successfully in therapeutic and prophylactic animal models to treat many different fungal pathogens, significantly increasing survival and reducing tissue fungal burden.
Collapse
Affiliation(s)
- Jill Adler-Moore
- Department of Biological Sciences, California State Polytechnic University, Pomona
| | - Russell E Lewis
- Unit of Infectious Diseases, Policlinico Sant'Orsola-Malpighi, Department of Medical Sciences and Surgery, University of Bologna, Italy
| | - Roger J M Brüggemann
- Department of Pharmacy, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Bart J A Rijnders
- Department of Internal Medicine, Section of Infectious Diseases, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Andreas H Groll
- Infectious Disease Research Program, Department of Pediatric Hematology and Oncology and Center for Bone Marrow Transplantation, University Children's Hospital Muenster, Germany
| | - Thomas J Walsh
- Departments of Medicine, Pediatrics, and Microbiology & Immunology, Weill Cornell Medicine of Cornell University, New York, New York
| |
Collapse
|
6
|
AL-Khikani FHO. Amphotericin B from antifungal to antiviral therapy: promising modern therapeutic branch. RESEARCH RESULTS IN PHARMACOLOGY 2020. [DOI: 10.3897/rrpharmacology.6.53649] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Introduction: Amphotericin B (AmB) which belongs to the polyene group has a wide spectrum in vitro and in vivo antimicrobial activity against fungi and parasites, but resistance to AmB is rare despite extensive use.
Material and methods: Atotal of 2530 articles were investigated in PubMed (n = 1525), Medline (n = 705), and Google Scholar (n = 300). From 2530 articles, only 61 studies were included in this review. All the short and full articles were searched that were scheduled to be published until April 2020.
Results: After its discovery, AmB has been one of the most common first-line choices in treating systemic fungal infection for over seven decades from its discovery. Recently, some studies have focused on the potential antimicrobial action of AmB against some enveloped and non-enveloped viruses, such as human immunodeficiency virus, Japanese encephalitis virus, herpes simplex virus, and Rubella virus.
Discussion: Among the invading pathogens, viruses constitute the most common ones,Due to the continuous spreading of viral infections with the rise in death numbers, new therapeutics development is urgent, as in general, some lethal viruses have no specific antiviral drugs or vaccines. So, this review may serve as an impetus for researchers working in the field of medical microbiology, vaccination, and antiviral drug design by discussing the most recent information about the antiviral action of AmB, as well as trying to provide a deeper understanding of major properties, mechanisms of action, immune system responses, and antimicrobial efficiency of AmB.
Conclusion: Since AmB is expected to alter the structure of the viral envelope, membrane integrity of cells, and internal cellular organelles, besides its other unique properties, such as host immunomodulatory effects, this review suggested that AmB as an effective anti-fungi drug may hold the promise of formulating a novel therapeutic option to treat many dangerous viruses, including those for treating which there are no active drugs or vaccines.
Collapse
|
7
|
Gandra RM, McCarron P, Viganor L, Fernandes MF, Kavanagh K, McCann M, Branquinha MH, Santos ALS, Howe O, Devereux M. In vivo Activity of Copper(II), Manganese(II), and Silver(I) 1,10-Phenanthroline Chelates Against Candida haemulonii Using the Galleria mellonella Model. Front Microbiol 2020; 11:470. [PMID: 32265890 PMCID: PMC7105610 DOI: 10.3389/fmicb.2020.00470] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 03/04/2020] [Indexed: 12/19/2022] Open
Abstract
Candida haemulonii is an emerging opportunistic pathogen resistant to most antifungal drugs currently used in clinical arena. Metal complexes containing 1,10-phenanthroline (phen) chelating ligands have well-established anti-Candida activity against different medically relevant species. This study utilized larvae of Galleria mellonella, a widely used model of in vivo infection, to examine C. haemulonii infection characteristics in response to different copper(II), manganese(II), and silver(I) chelates containing phen, which had demonstrated potent anti-C. haemulonii activity in a previous study. The results showed that C. haemulonii virulence was influenced by inoculum size and incubation temperature, and the host G. mellonella immune response was triggered in an inoculum-dependent manner reflected by the number of circulating immune cells (hemocytes) and observance of larval melanization process. All test chelates were non-toxic to the host in concentrations up to 10 μg/larva. The complexes also affected the G. mellonella immune system, affecting the hemocyte number and the expression of genes encoding antifungal and immune-related peptides (e.g., inducible metalloproteinase inhibitor protein, transferrin, galiomycin, and gallerimycin). Except for [Ag2(3,6,9-tdda)(phen)4].EtOH (3,6,9-tddaH2 = 3,6,9-trioxoundecanedioic acid), all chelates were capable of affecting the fungal burden of infected larvae and the virulence of C. haemulonii in a dose-dependent manner. This work shows that copper(II), manganese(II), and silver(I) chelates containing phen with anti-C. haemulonii activity are capable of (i) inhibiting fungal proliferation during in vivo infection, (ii) priming an immune response in the G. mellonella host and (iii) affecting C. haemulonii virulence.
Collapse
Affiliation(s)
- Rafael M Gandra
- Laboratório de Estudos Avançados de Microrganismos Emergentes e Resistentes, Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Instituto de Química, Programa de Pós-Graduação em Bioquímica, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Centre for Biomimetic and Therapeutic Research, Focas Research Institute, Technological University Dublin, Dublin, Ireland
| | - Pauraic McCarron
- Centre for Biomimetic and Therapeutic Research, Focas Research Institute, Technological University Dublin, Dublin, Ireland
| | - Livia Viganor
- Laboratório de Estudos Avançados de Microrganismos Emergentes e Resistentes, Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Centre for Biomimetic and Therapeutic Research, Focas Research Institute, Technological University Dublin, Dublin, Ireland
| | - Mariana Farias Fernandes
- Laboratório de Estudos Avançados de Microrganismos Emergentes e Resistentes, Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Kevin Kavanagh
- Department of Biology, Maynooth University, National University of Ireland, Maynooth, Ireland
| | - Malachy McCann
- Department of Chemistry, Maynooth University, National University of Ireland, Maynooth, Ireland
| | - Marta H Branquinha
- Laboratório de Estudos Avançados de Microrganismos Emergentes e Resistentes, Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - André L S Santos
- Laboratório de Estudos Avançados de Microrganismos Emergentes e Resistentes, Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Instituto de Química, Programa de Pós-Graduação em Bioquímica, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Orla Howe
- Centre for Biomimetic and Therapeutic Research, Focas Research Institute, Technological University Dublin, Dublin, Ireland.,School of Biological & Health Sciences, Technological University Dublin, Dublin, Ireland
| | - Michael Devereux
- Centre for Biomimetic and Therapeutic Research, Focas Research Institute, Technological University Dublin, Dublin, Ireland
| |
Collapse
|
8
|
Obayes AL-Khikani F, Ayit A. Prospects in Immunomodulatory activity of Amphotericin B in viral infection: Promising developing therapeutic branch. JOURNAL OF CURRENT RESEARCH IN SCIENTIFIC MEDICINE 2020. [DOI: 10.4103/jcrsm.jcrsm_29_20] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
9
|
Decker C, Wurster S, Lazariotou M, Hellmann AM, Einsele H, Ullmann AJ, Löffler J. Analysis of the in vitro activity of human neutrophils against Aspergillus fumigatus in presence of antifungal and immunosuppressive agents. Med Mycol 2019; 56:514-519. [PMID: 29420763 DOI: 10.1093/mmy/myx069] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Accepted: 09/15/2017] [Indexed: 12/22/2022] Open
Abstract
Neutrophils are essential in the first line defense against moulds. This in vitro study assessed different neutrophil effector mechanisms in the presence of clinically relevant antifungal and immunosuppressive agents. Therapeutic concentrations of liposomal amphotericin B led to reduced IL-8 and oxidative burst response to the synthetic stimulus PMA, whereas no major alterations of oxidative burst, phagocytosis, or cytokine response to germinated stages of Aspergillus fumigatus and no supra-additive effects of antifungal and immunosuppressive drugs were observed. Conventional and liposomal amphotericin B as well as voriconazole, however, led to reduced neutrophil extracellular trap formation in response to A. fumigatus germ tubes.
Collapse
Affiliation(s)
- Christina Decker
- University Hospital of Wuerzburg, Department of Internal Medicine II, Wuerzburg
| | - Sebastian Wurster
- University Hospital of Wuerzburg, Department of Internal Medicine II, Wuerzburg
| | - Maria Lazariotou
- University Hospital of Wuerzburg, Department of Internal Medicine II, Wuerzburg
| | - Anna-Maria Hellmann
- University Hospital of Wuerzburg, Department of Internal Medicine II, Wuerzburg
| | - Hermann Einsele
- University Hospital of Wuerzburg, Department of Internal Medicine II, Wuerzburg
| | - Andrew J Ullmann
- University Hospital of Wuerzburg, Department of Internal Medicine II, Wuerzburg
| | - Jürgen Löffler
- University Hospital of Wuerzburg, Department of Internal Medicine II, Wuerzburg
| |
Collapse
|
10
|
Dellière S, Guery R, Candon S, Rammaert B, Aguilar C, Lanternier F, Chatenoud L, Lortholary O. Understanding Pathogenesis and Care Challenges of Immune Reconstitution Inflammatory Syndrome in Fungal Infections. J Fungi (Basel) 2018; 4:E139. [PMID: 30562960 PMCID: PMC6308948 DOI: 10.3390/jof4040139] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 12/14/2018] [Accepted: 12/15/2018] [Indexed: 12/16/2022] Open
Abstract
Immune deficiency of diverse etiology, including human immunodeficiency virus (HIV), antineoplastic agents, immunosuppressive agents used in solid organ recipients, immunomodulatory therapy, and other biologics, all promote invasive fungal infections. Subsequent voluntary or unintended immune recovery may induce an exaggerated inflammatory response defining immune reconstitution inflammatory syndrome (IRIS), which causes significant mortality and morbidity. Fungal-associated IRIS raises several diagnostic and management issues. Mostly studied with Cryptococcus, it has also been described with other major fungi implicated in human invasive fungal infections, such as Pneumocystis, Aspergillus, Candida, and Histoplasma. Furthermore, the understanding of IRIS pathogenesis remains in its infancy. This review summarizes current knowledge regarding the clinical characteristics of IRIS depending on fungal species and existing strategies to predict, prevent, and treat IRIS in this patient population, and tries to propose a common immunological background to fungal IRIS.
Collapse
Affiliation(s)
- Sarah Dellière
- Medical School, Paris-Descartes University, APHP, Necker-Enfants Malades Hospital, Infectious Disease Center Necker-Pasteur, IHU Imagine, 75015 Paris, France.
| | - Romain Guery
- Medical School, Paris-Descartes University, APHP, Necker-Enfants Malades Hospital, Infectious Disease Center Necker-Pasteur, IHU Imagine, 75015 Paris, France.
| | - Sophie Candon
- Medical School, Paris-Descartes University, INSERM U1151-CNRS UMR 8253APHP, Necker-Enfants Malades Hospital, APHP, Clinical Immunology, 75015 Paris, France.
| | - Blandine Rammaert
- Medical School, Poitiers University, Poitiers, France; Poitiers University Hospital, Infectious Disease Unit, Poitiers, France; INSERM U1070, 86022 Poitiers, France.
| | - Claire Aguilar
- Medical School, Paris-Descartes University, APHP, Necker-Enfants Malades Hospital, Infectious Disease Center Necker-Pasteur, IHU Imagine, 75015 Paris, France.
| | - Fanny Lanternier
- Medical School, Paris-Descartes University, APHP, Necker-Enfants Malades Hospital, Infectious Disease Center Necker-Pasteur, IHU Imagine, 75015 Paris, France.
- Pasteur Institute, Molecular Mycology Unit, National Reference Center for Invasive Fungal Disease and Antifungals, CNRS UMR 2000, 75015 Paris, France.
| | - Lucienne Chatenoud
- Medical School, Paris-Descartes University, INSERM U1151-CNRS UMR 8253APHP, Necker-Enfants Malades Hospital, APHP, Clinical Immunology, 75015 Paris, France.
| | - Olivier Lortholary
- Medical School, Paris-Descartes University, APHP, Necker-Enfants Malades Hospital, Infectious Disease Center Necker-Pasteur, IHU Imagine, 75015 Paris, France.
- Pasteur Institute, Molecular Mycology Unit, National Reference Center for Invasive Fungal Disease and Antifungals, CNRS UMR 2000, 75015 Paris, France.
| |
Collapse
|
11
|
Ben-Ami R. Angiogenesis at the mold-host interface: a potential key to understanding and treating invasive aspergillosis. Future Microbiol 2014; 8:1453-62. [PMID: 24199803 DOI: 10.2217/fmb.13.114] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Invasive aspergillosis (IA) in neutropenic patients is characterized by angioinvasion, intravascular thrombosis and tissue infarction, features that lead to sequestration of infected tissue and impaired fungal clearance. Recent research has shown that host angiogenesis, the homeostatic compensatory response to tissue hypoxia, is downregulated by Aspergillus fumigatus secondary metabolites. A. fumigatus metabolites inhibit multiple key angiogenic mediators, notably basic FGF, VEGF and their respective receptors. Moreover, repletion of basic FGF and VEGF enhances angiogenesis at the site of infection, induces trafficking of polymorphonuclear leukocytes into fungal-infected tissue and enhances antifungal drug activity. This review summarizes the emerging roles of vasculopathy and angiogenesis in the pathogenesis of IA, emphasizing the importance of the underlying mode of immunosuppression. Modulation of angiogenesis is a potential target for novel therapeutic strategies against IA.
Collapse
Affiliation(s)
- Ronen Ben-Ami
- Infectious Diseases Unit, Tel Aviv Medical Center & the Sackler School of Medicine, Tel Aviv University, Israel.
| |
Collapse
|
12
|
Kuyucu N. Amphotericin B use in children: conventional and lipid-based formulations. Expert Rev Anti Infect Ther 2014; 9:357-67. [DOI: 10.1586/eri.11.5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
13
|
Van Waeyenberghe L, Baré J, Pasmans F, Claeys M, Bert W, Haesebrouck F, Houf K, Martel A. Interaction of Aspergillus fumigatus conidia with Acanthamoeba castellanii parallels macrophage-fungus interactions. ENVIRONMENTAL MICROBIOLOGY REPORTS 2013; 5:819-824. [PMID: 24249290 DOI: 10.1111/1758-2229.12082] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2012] [Accepted: 06/22/2013] [Indexed: 06/02/2023]
Abstract
Aspergillus fumigatus and free-living amoebae are common inhabitants of soil. Mechanisms of A. fumigatus to circumvent the amoeba's digestion may facilitate overcoming the vertebrate macrophage defence mechanisms. We performed co-culture experiments using A. fumigatus conidia and the amoeba Acanthamoeba castellanii. Approximately 25% of the amoebae ingested A. fumigatus conidia after 1 h of contact. During intra-amoebal passage, part of the ingested conidia was able to escape the food vacuole and to germinate inside the cytoplasm of A. castellanii. Fungal release into the extra-protozoan environment by exocytosis of conidia or by germination was observed with light and transmission electron microscopy. These processes resulted in structural changes in A. castellanii, leading to amoebal permeabilization without cell lysis. In conclusion, A. castellanii internalizes A. fumigatus conidia, resulting in fungal intracellular germination and subsequent amoebal death. As such, this interaction highly resembles that of A. fumigatus with mammalian and avian macrophages. This suggests that A. fumigatus virulence mechanisms to evade macrophage killing may be acquired by co-evolutionary interactions among A. fumigatus and environmental amoebae.
Collapse
Affiliation(s)
- Lieven Van Waeyenberghe
- Department of Pathology, Bacteriology and Avian Diseases, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, B-9820, Merelbeke, Belgium
| | | | | | | | | | | | | | | |
Collapse
|
14
|
Ben-Ami R, Albert ND, Lewis RE, Kontoyiannis DP. Proangiogenic growth factors potentiate in situ angiogenesis and enhance antifungal drug activity in murine invasive aspergillosis. J Infect Dis 2013; 207:1066-74. [PMID: 23303813 DOI: 10.1093/infdis/jis940] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
In invasive pulmonary aspergillosis, direct invasion and occlusion of pulmonary vasculature by Aspergillus hyphae causes tissue hypoxia, which is enhanced by secreted fungal metabolites that downregulate compensatory angiogenic signaling pathways. We assessed the effects of basic fibroblast growth factor (bFGF) and vascular endothelial growth factor (VEGF) on survival rates, fungal burden, and in situ angiogenesis in a murine invasive pulmonary aspergillosis model. bFGF and VEGF monotherapy significantly increased survival rates and potentiated the activity of amphotericin B. bFGF-containing regimens were associated with reduced tissue fungal burdens. bFGF and VEGF reversed the antiangiogenic activity of Aspergillus fumigatus; however, VEGF induced the formation of immature neovessels, providing an explanation for its lesser efficacy. Treatment with bFGF plus amphotericin B was associated with neutrophil influx into Aspergillus-infected pulmonary tissue, suggesting that this combination limits fungal growth through neutrophil trafficking. Vasculogenic pathways are unexplored targets for the treatment of invasive pulmonary aspergillosis and may potentiate both innate immunity and antifungal drug activity against A. fumigatus.
Collapse
Affiliation(s)
- Ronen Ben-Ami
- Infectious Diseases Unit, Tel Aviv Sourasky Medical Center, Israel.
| | | | | | | |
Collapse
|
15
|
Lewis RE, Viale P. Update on Amphotericin B Pharmacology and Dosing for Common Systemic Mycoses. CURRENT FUNGAL INFECTION REPORTS 2012. [DOI: 10.1007/s12281-012-0107-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
16
|
Mesa-Arango AC, Scorzoni L, Zaragoza O. It only takes one to do many jobs: Amphotericin B as antifungal and immunomodulatory drug. Front Microbiol 2012; 3:286. [PMID: 23024638 PMCID: PMC3441194 DOI: 10.3389/fmicb.2012.00286] [Citation(s) in RCA: 170] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2012] [Accepted: 07/21/2012] [Indexed: 11/26/2022] Open
Abstract
“Amphotericin B acts through pore formation at the cell membrane after binding to ergosterol” is an accepted dogma about the action mechanism of this antifungal, and this sentence is widely found in the literature. But after 60 years of investigation, the action mechanism of Amphotericin B is not fully elucidated. Amphotericin B is a polyene substance that is one of the most effective drugs for the treatment of fungal and parasite infections. As stated above, the first mechanism of action described was pore formation after binding to the ergosterol present in the membrane. But it has also been demonstrated that AmB induces oxidative damage in the cells. Moreover, amphotericin B modulates the immune system, and this activity has been related to the protective effect of the molecule, but also to its toxicity in the host. This review tries to provide a general overview of the main aspects of this molecule, and highlight the multiple effects that this molecule has on both the fungal and host cells.
Collapse
Affiliation(s)
- Ana C Mesa-Arango
- Mycology Reference Laboratory, National Centre for Microbiology, Instituto de Salud Carlos III Majadahonda, Madrid, Spain ; Group of Investigative Dermatology, University of Antioquia Medellín, Colombia
| | | | | |
Collapse
|
17
|
Omran NEE, Allam NG. Screening of microbial contamination and antimicrobial activity of sea cucumber Holothuria polii. Toxicol Ind Health 2012; 29:944-54. [DOI: 10.1177/0748233712448116] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Microbiological studies were carried out on microbial contamination and antimicrobial activity of sea cucumber Holothuria polii collected from Mediterranean Sea at Abu-kir shore of Alexandria, Egypt. The obtained results revealed the presence of isolates of five human Gram-negative pathogenic bacteria, representing five genera were identified to species level, including, Esherichia coli, Klebsiella pneumoniae, Pseudomonas aeruginosa, Salmonella sp. and Shigella sp. In addition, an yeast Candida albicans was isolated. The pathogenic bacteria were identified using API 20E strip system (BioMereux). All collected H. polii specimens were healthy with no external signs of infection. Histopathological study of the tegument, intestine and gonads showed no abnormal changes. The antimicrobial activity of two tegumental ethanol extracts (A and B, differ in the method of dehydration) were tested against wide range of pathogenic bacteria and fungi, including intestinal, skin and nosocomial pathogens and one plant fungal pathogen. The results revealed a remarkable antifungal activity of the extract B at 2.5 mg/ml MIC90, especially on Aspergillus niger, Scloretium sp, C. albicans, Aspergillus flavus and Malassezia furfur, and limited antibacterial activity against Gram-negative bacteria ( Salmonella choleraesuis ATCC 14028 and Aeromonas hydrophila). The domain of bacterial and limited fungal contamination confirms the results that showed strong antifungal activity of investigated extract.
Collapse
Affiliation(s)
- Nahla EE Omran
- Invertebrate Unit, Department of Zoology, Faculty of Science, Tanta University, Tanta, Egypt
| | - Nanis G Allam
- Microbiology Unit, Department of Botany, Faculty of Science, Tanta University, Tanta, Egypt
| |
Collapse
|
18
|
Simitsopoulou M, Roilides E, Walsh TJ. Immunomodulatory properties of antifungal agents on phagocytic cells. Immunol Invest 2012; 40:809-24. [PMID: 21985307 DOI: 10.3109/08820139.2011.615877] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Phagocytic cells, particularly neutrophils and monocytes/macrophages, are the first line and the most effective form of innate host defence against pathogenic fungi. During antifungal therapy these phagocytic cells are also exposed to antifungal agents. In the phagocyte-fungus-antifungal agent interplay, drugs may directly interact with phagocytes through specific pattern recognition receptors, leading to altered antifungal activities. Antifungal agents, through modulation of fungal virulence, may initiate different immune response programs in the phagocytes, leading to antifungal synergism/antagonism or up-regulation of gene expression for a pro-inflammatory response. Additionally, indirect modulation of phagocyte behavior by pretreatment of neutrophils, monocytes, and macrophages with cytokines and exposure to antifungal agents have shown promising findings for combined drug-cytokine therapy that may improve treatment of life-threatening fungal diseases. In this review, we discuss the main in vitro and in vivo immunomodulatory effects of antifungal agents on phagocytes in response to pathogenic fungi, as well as we address underlying immunopharmacologic mechanisms and their potential impact on clinical outcome.
Collapse
Affiliation(s)
- Maria Simitsopoulou
- Laboratory of Infectious Diseases, 3rd Department of Paediatrics, School of Medicine, Aristotle University, Hippokration Hospital, Thessaloniki, Greece
| | | | | |
Collapse
|
19
|
The initial 96 hours of invasive pulmonary aspergillosis: histopathology, comparative kinetics of galactomannan and (1->3) β-d-glucan and consequences of delayed antifungal therapy. Antimicrob Agents Chemother 2010; 54:4879-86. [PMID: 20713673 DOI: 10.1128/aac.00673-10] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Acute invasive pulmonary aspergillosis is a rapidly progressive and frequently lethal infection. Relatively little is known about early events in the pathogenesis and relationship between the cell wall biomarkers galactomannan and (1→3)-β-d-glucan. The consequences of delayed antifungal therapy are also poorly defined. A persistently neutropenic rabbit model of invasive pulmonary aspergillosis was used to describe the histopathology of early invasive pulmonary aspergillosis and the kinetics of galactomannan and (1→3)-β-d-glucan. The time course of both molecules was mathematically modeled by using a population methodology, and Monte Carlo simulations were performed. The effect of progressive delay in the administration of amphotericin B deoxycholate 1 mg/kg at 24, 48, 72, and 96 h postinoculation on fungal burden, lung weight, pulmonary infarct score, and survival was determined. Histopathology showed phagocytosis of conidia by pulmonary alveolar macrophages at 4 h postinoculation. At 12 to 24 h, there was a progressive focal inflammatory response with conidial germination and hyphal extension. Subsequently, hyphae invaded into the contiguous lung. Galactomannan and (1→3)-β-d-glucan had similar trajectories, and both exhibited considerable interindividual variability, which was reflected in Monte Carlo simulations. Concentrations of both molecules began to rise <24 h postinoculation before pulmonary hemorrhagic infarction was present. Delays of 72 and 96 h in the administration of amphotericin B resulted in fungal burdens and lung weights that were indistinguishable from those of controls, respectively. Galactomannan and (1→3)-β-d-glucan have similar kinetics and are comparable biomarkers of early invasive pulmonary aspergillosis. Antifungal treatment at ≥48 h postinoculation is associated with suboptimal therapeutic outcomes.
Collapse
|
20
|
Perkhofer S, Trappl K, Nussbaumer W, Dierich MP, Lass-Flörl C. Potential synergistic activity of antimycotic substances in combination with human platelets against Aspergillus fumigatus. J Antimicrob Chemother 2010; 65:1309-11. [PMID: 20378674 DOI: 10.1093/jac/dkq111] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
21
|
Leventakos K, Ben-Ami R, Lewis RE, Kontoyiannis DP. Immunomodulating effects of antifungal therapy. CURRENT FUNGAL INFECTION REPORTS 2009. [DOI: 10.1007/s12281-009-0034-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
22
|
Pagano L, Caira M, Candoni A, Offidani M, Martino B, Specchia G, Pastore D, Stanzani M, Cattaneo C, Fanci R, Caramatti C, Rossini F, Luppi M, Potenza L, Ferrara F, Mitra ME, Fadda RM, Invernizzi R, Aloisi T, Picardi M, Bonini A, Vacca A, Chierichini A, Melillo L, de Waure C, Fianchi L, Riva M, Leone G, Aversa F, Nosari A. Invasive aspergillosis in patients with acute myeloid leukemia: a SEIFEM-2008 registry study. Haematologica 2009; 95:644-50. [PMID: 19850903 DOI: 10.3324/haematol.2009.012054] [Citation(s) in RCA: 243] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND The aim of this study was to evaluate prognostic factors, treatments and outcome of invasive aspergillosis in patients with acute myeloid leukemia based on data collected in a registry. DESIGN AND METHODS The registry, which was activated in 2004 and closed in 2007, collected data on patients with acute myeloid leukemia, admitted to 21 hematologic divisions in tertiary care centers or university hospitals in Italy, who developed proven or probable invasive aspergillosis. RESULTS One hundred and forty cases of invasive aspergillosis were collected, with most cases occurring during the period of post-induction aplasia, the highest risk phase in acute myeloid leukemia. The mortality rate attributable to invasive aspergillosis was 27%, confirming previous reports of a downward trend in this rate. Univariate and multivariate analyses revealed that the stage of acute myeloid leukemia and the duration of, and recovery from, neutropenia were independent prognostic factors. We analyzed outcomes after treatment with the three most frequently used drugs (liposomal amphotericin B, caspofungin, voriconazole). No differences emerged in survival at day 120 or in the overall response rate which was 71%, ranging from 61% with caspofungin to 84% with voriconazole. CONCLUSIONS Our series confirms the downward trend in mortality rates reported in previous series, with all new drugs providing similar survival and response rates. Recovery from neutropenia and disease stage are crucial prognostic factors. Efficacious antifungal drugs bridge the period of maximum risk due to poor hematologic and immunological reconstitution.
Collapse
Affiliation(s)
- Livio Pagano
- M.D. Istituto di Ematologia, Università Cattolica del Sacro Cuore, Largo Francesco Vito, 1 I-00168 Roma, Italia.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Moen MD, Lyseng-Williamson KA, Scott LJ. Liposomal amphotericin B: a review of its use as empirical therapy in febrile neutropenia and in the treatment of invasive fungal infections. Drugs 2009; 69:361-92. [PMID: 19275278 DOI: 10.2165/00003495-200969030-00010] [Citation(s) in RCA: 217] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Liposomal amphotericin B (AmBisome) is a lipid-associated formulation of the broad-spectrum polyene antifungal agent amphotericin B. It is active against clinically relevant yeasts and moulds, including Candida spp., Aspergillus spp. and filamentous moulds such as Zygomycetes, and is approved for the treatment of invasive fungal infections in many countries worldwide. It was developed to improve the tolerability profile of amphotericin B deoxycholate, which was for many decades considered the gold standard of antifungal treatment, despite being associated with infusion-related events and nephrotoxicity. In well controlled trials, liposomal amphotericin B had similar efficacy to amphotericin B deoxycholate and amphotericin B lipid complex as empirical therapy in adult and paediatric patients with febrile neutropenia. In addition, caspofungin was noninferior to liposomal amphotericin B as empirical therapy in adult patients with febrile neutropenia. For the treatment of confirmed invasive fungal infections, liposomal amphotericin B was more effective than amphotericin B deoxycholate treatment in patients with disseminated histoplasmosis and AIDS, and was noninferior to amphotericin B deoxycholate in patients with acute cryptococcal meningitis and AIDS. In adults, micafungin was shown to be noninferior to liposomal amphotericin B for the treatment of candidaemia and invasive candidiasis. Data from animal studies suggested that higher dosages of liposomal amphotericin B might improve efficacy; however, in the AmBiLoad trial in patients with invasive mould infection, there was no statistical difference in efficacy between the standard dosage of liposomal amphotericin B 3 mg/kg/day and a higher 10 mg/kg/day dosage, although the standard dosage was better tolerated. Despite being associated with fewer infusion-related adverse events and less nephrotoxicity than amphotericin B deoxycholate and amphotericin B lipid complex, liposomal amphotericin B use is still limited to some extent by these adverse events. Both echinocandins were better tolerated than liposomal amphotericin B. The cost of liposomal amphotericin B therapy may also restrict its use, but further pharmacoeconomic studies are required to fully define its cost effectiveness compared with other antifungal agents. Based on comparative data from well controlled trials, extensive clinical experience and its broad spectrum of activity, liposomal amphotericin B remains a first-line option for empirical therapy in patients with febrile neutropenia and in those with disseminated histoplasmosis, and is an option for the treatment of AIDS-associated cryptococcal meningitis, and for invasive Candida spp. or Aspergillus spp. infections. Amphotericin B, a macrocyclic, polyene antifungal agent, is thought to act by binding to ergosterol, the principal sterol in fungal cell membranes and Leishmania cells. This results in a change in membrane permeability, causing metabolic disturbance, leakage of small molecules and, as a consequence, cell death. In vitro and in vivo studies have shown that liposomal amphotericin B remains closely associated with the liposomes in the circulation, thereby reducing the potential for nephrotoxicity and infusion-related toxicity associated with conventional amphotericin B. Amphotericin B shows very good in vitro activity against a broad spectrum of clinically relevant fungal isolates, including most strains of Candida spp. and Aspergillus spp., and other filamentous fungi such as Zygomycetes. Liposomal amphotericin B has proven effective in various animal models of fungal infections, including those for candidiasis, aspergillosis, fusariosis and zygomycosis. Liposomal amphotericin B also shows immunomodulatory effects, although the mechanisms involved are not fully understood, and differ from those of amphotericin B deoxycholate and amphotericin B colloidal dispersion. In adult patients with febrile neutropenia, intravenous liposomal amphotericin B has nonlinear pharmacokinetics, with higher than dose-proportional increases in exposure being consistent with reticuloendothelial saturation and redistribution of amphotericin B in the plasma compartment. Liposomal amphotericin B is rapidly and extensively distributed after single and multiple doses, with steady-state concentrations of amphotericin B attained within 4 days and no clinically relevant accumulation of the drug following multiple doses of 1-7.5 mg/kg/day. In autopsy tissue, the highest concentrations of the drug were found in the liver and spleen, followed by the kidney, lung, myocardium and brain tissue. Elimination of liposomal amphotericin B, like that of amphotericin B deoxycholate, is poorly understood; its route of metabolism is not known and its excretion has not been studied. The terminal elimination half-life is about 7 hours. No dosage adjustment is required based on age or renal impairment. In several randomized, double-blind trials (n = 73-1095) in adult and/or paediatric patients, liposomal amphotericin B was effective as empirical therapy or as treatment for confirmed invasive fungal infections, including invasive candidiasis, candidaemia, invasive mould infection (mainly aspergillosis), histoplasmosis and cryptococcal meningitis. All agents were administered as an intravenous infusion; the typical dosage for liposomal amphotericin B was 3 mg/kg/day. Treatment was generally given for 1-2 weeks. Participants in trials evaluating empirical therapy had neutropenia and a persistent fever despite antibacterial treatment and had received chemotherapy or undergone haematopoietic stem cell transplantation. As empirical therapy in adult and paediatric patients, liposomal amphotericin B appeared to be as effective as amphotericin B deoxycholate (approximately 50% of patients in each group achieved treatment success) or amphotericin B lipid complex (approximately 40% of liposomal amphotericin B recipients experienced treatment success). Of note, in the first trial, results of the statistical test to determine equivalence between treatments were not reported. In the second trial, efficacy was assessed as an 'other' endpoint. In another trial, caspofungin was shown to be noninferior to liposomal amphotericin B, with approximately one-third of patients in each group experiencing treatment success. Liposomal amphotericin B was significantly more effective than amphotericin B deoxycholate for the treatment of moderate to severe disseminated histoplasmosis in patients with AIDS, with 88% and 64% of patients, respectively, having a successful response. Liposomal amphotericin B was noninferior to amphotericin B deoxycholate for the treatment of cryptococcal meningitis in terms of mycological success. Micafungin therapy was shown to be noninferior to liposomal amphotericin B for the treatment of adult patients with candidaemia or invasive candidiasis. In a substudy in paediatric patients, which was not powered to determine noninferiority, liposomal amphotericin B was as effective as micafungin for the treatment of candidaemia or invasive candidiasis. In this patient population, within each trial, 90% of adult patients and approximately three-quarters of paediatric patients in both treatment groups experienced a successful response. In patients with invasive mould infection (mainly aspergillosis), there was no difference in efficacy between a higher dosage of liposomal amphotericin B (10 mg/kg/day) and the standard dosage (3 mg/kg/day), with 46% and 50% of patients experiencing a favourable overall response. In well designed clinical trials, liposomal amphotericin B was generally at least as well tolerated as other lipid-associated formulations of amphotericin B and better tolerated than amphotericin B deoxycholate in adult and paediatric patients. Compared with other amphotericin B formulations, liposomal amphotericin B treatment was associated with a lower incidence of infusion-related adverse events and nephrotoxicity. A higher than recommended dosage of liposomal amphotericin B (10 mg/kg/day) was associated with an increased incidence of nephrotoxicity compared with the standard dosage (3 mg/kg/day), although the incidence of infusion-related reactions did not differ between treatment groups. In general, liposomal amphotericin B treatment was not as well tolerated as echinocandin therapy in well designed clinical trials. As empirical therapy or for the treatment of confirmed invasive fungal infections in adult patients, liposomal amphotericin B recipients experienced more infusion-related events and nephrotoxicity than caspofungin or micafungin recipients. There was no difference in the incidence of these adverse events between the liposomal amphotericin B and micafungin groups in a study in paediatric patients.
Collapse
Affiliation(s)
- Marit D Moen
- Wolters Kluwer Health
- Adis, Auckland, New Zealand.
| | | | | |
Collapse
|
24
|
Lewis RE, Kontoyiannis DP. Invasive aspergillosis in glucocorticoid-treated patients. Med Mycol 2009; 47 Suppl 1:S271-81. [DOI: 10.1080/13693780802227159] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
|
25
|
Ben‐Ami R, Lewis R, Kontoyiannis D. Immunocompromised Hosts: Immunopharmacology of Modern Antifungals. Clin Infect Dis 2008; 47:226-35. [DOI: 10.1086/589290] [Citation(s) in RCA: 109] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
|
26
|
Simitsopoulou M, Roilides E, Maloukou A, Gil-Lamaignere C, Walsh TJ. Interaction of amphotericin B lipid formulations and triazoles with human polymorphonuclear leucocytes for antifungal activity against Zygomycetes. Mycoses 2008; 51:147-54. [DOI: 10.1111/j.1439-0507.2007.01457.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
27
|
Perkhofer S, Speth C, Dierich MP, Lass-Flörl C. In vitro determination of phagocytosis and intracellular killing of Aspergillus species by mononuclear phagocytes. Mycopathologia 2007; 163:303-7. [PMID: 17458710 DOI: 10.1007/s11046-007-9013-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2007] [Accepted: 03/21/2007] [Indexed: 10/23/2022]
Abstract
We investigated phagocytosis and intracellular killing of clinical and environmental isolates of Aspergillus spp. by human monocyte-derived macrophages (MDMs). Serial pathogens such as Aspergillus fumigatus, Aspergillus flavus and Aspergillus terreus were examined with a microbiological assay. Phagocytosis for resting conidia of Aspergillus spp. was similar for all isolates tested. During 30 min of incubation phagocytosis ranged from 49.9% to 85.5% for clinical isolates and from 40.3% to 87.1% for environmental isolates. MDMs killed A. fumigatus, A. flavus and A. terreus conidia after ingestion for 120 min, as shown by a decrease in colony forming units (cfu) count of intracellular fungi. The killing index for all isolates of Aspergillus spp., ranged from 12.1 +/- 1.1% to 90.3 +/- 10.4%; isolate-dependent (P < 0.01) differences against the fungicidal action of MDMs were observed. In conclusion, significant differences were noted for killing indices between several strains of Aspergillus spp. whereas phagocytosis was similar for all isolates tested in vitro. No differences were observed within environmental and clinical isolates.
Collapse
Affiliation(s)
- Susanne Perkhofer
- Department of Hygiene, Microbiology and Social Medicine, Medical University Innsbruck, Fritz Pregl Strasse 3, 6020, Innsbruck, Austria.
| | | | | | | |
Collapse
|
28
|
Dotis J, Simitsopoulou M, Dalakiouridou M, Konstantinou T, Taparkou A, Kanakoudi-Tsakalidou F, Walsh TJ, Roilides E. Effects of lipid formulations of amphotericin B on activity of human monocytes against Aspergillus fumigatus. Antimicrob Agents Chemother 2006; 50:868-73. [PMID: 16495244 PMCID: PMC1426430 DOI: 10.1128/aac.50.3.868-873.2006] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The immunomodulatory effects of liposomal amphotericin B (LAMB), amphotericin B lipid complex, and amphotericin B colloidal dispersion (ABCD) on antifungal activity of human monocytes (MNCs), an important component of antifungal host defense, against Aspergillus fumigatus were compared to those of deoxycholate amphotericin B (DAMB). MNCs from healthy volunteers were incubated with 1 or 5 microg/ml DAMB and 5 or 25 microg/ml lipid formulations for 22 h. Drug-pretreated or untreated MNCs were then washed and assayed for the following: (i) activity against A. fumigatus hyphae by XTT assay at MNC:hypha ratios of 10:1 and 20:1; (ii) production of superoxide anion (O2-) from MNCs in response to hyphae by cytochrome c reduction; (iii) production of hydrogen peroxide (H2O2) and H2O2-dependent intracellular intermediates (DIIs), such as OH- and HOCl, from MNCs in response to A. fumigatus culture supernatant by flow cytometric measurement of dihydrorhodamine-1,2,3 oxidation. With the exception of 1 microg/ml DAMB and 5 mug/ml LAMB or ABCD at 10:1, all amphotericin B formulations at both concentrations and MNC:hypha ratios enhanced MNC-induced damage of A. fumigatus hyphae compared to results with untreated cells (P < 0.01). While MNC O2- production upon hyphal challenge, an early event in oxidative burst, was not affected by the drugs, production of H2O2 and DIIs, late events, were significantly increased by all four drugs (P < 0.01). At clinically relevant concentrations, both conventional amphotericin B and its lipid formulations enhance antihyphal activity of MNCs against A. fumigatus in association with significant augmentation of H2O2 and DIIs but not O2-, further demonstrating the immunomodulatory antifungal activities of these agents.
Collapse
Affiliation(s)
- J Dotis
- Laboratory of Infectious Diseases, 3rd Department of Pediatrics, Aristotle University, Hippokration Hospital, Konstantinoupoleos 49, GR-546 42 Thessaloniki, Greece
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Roilides E, Lyman CA, Armstrong D, Stergiopoulou T, Petraitiene R, Walsh TJ. Deoxycholate amphotericin B and amphotericin B lipid complex exert additive antifungal activity in combination with pulmonary alveolar macrophages against Fusarium solani. Mycoses 2006; 49:109-13. [PMID: 16466443 DOI: 10.1111/j.1439-0507.2006.01202.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Fusarium spp. have emerged as important causes of invasive fungal infections in immunocompromised patients. Rabbit pulmonary alveolar macrophages (PAMs) exhibited fungicidal activity against conidia of Fusarium solani and achieved a time-dependent increase in killing. Neither deoxycholate amphotericin B (DAMB) nor amphotericin B lipid complex (ABLC) exerted a suppressive effect on PAMs by decreasing their conidiocidal activity against F. solani. On the contrary, at a concentration of 0.125 microg ml(-1), ABLC and, to a lesser degree, DAMB additively augmented the fungicidal activity of pulmonary alveolar macrophages against conidia of Fusarium solani.
Collapse
Affiliation(s)
- Emmanuel Roilides
- Immunocompromised Host Section, Pediatric Oncology Branch, National Cancer Institute, Bethesda, MA, USA
| | | | | | | | | | | |
Collapse
|
30
|
Takemoto K, Yamamoto Y, Ueda Y, Sumita Y, Yoshida K, Niki Y. Comparative study on the efficacy of AmBisome and Fungizone in a mouse model of pulmonary aspergillosis. J Antimicrob Chemother 2006; 57:724-31. [PMID: 16446374 DOI: 10.1093/jac/dkl005] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
OBJECTIVES The aim of this study was to evaluate the efficacy and tissue concentration of AmBisome and Fungizone in murine pulmonary aspergillosis, and to investigate the localization of AmBisome at the infection site. METHODS Mice were infected intratracheally with Aspergillus fumigatus. A single dose of each of the antifungals was administered intravenously 4 h after infection. The efficacy of the antifungal treatment was assessed by the pulmonary fungal burden at 20 h post-treatment and the survival time over 1 month. The pulmonary amphotericin B (AMB) concentration was measured until 48 h after administration. The distribution of AmBisome in the lung was evaluated using rhodamine-labelled AmBisome and an anti-AMB antibody. RESULTS AmBisome at a dose of > or =1 mg/kg significantly prolonged the survival time of infected mice compared with the control group. At the maximum tolerated dose, 10 mg/kg AmBisome exhibited greater efficacy than 1 mg/kg Fungizone in terms of increasing survival and reducing the fungal burden. The pulmonary AMB concentration of 10 mg/kg AmBisome was higher than that of 1 mg/kg Fungizone. Tissue distribution analysis showed that AmBisome was localized at the infection site in the lung, and this might explain the potent in vivo efficacy in this infection model. CONCLUSIONS AmBisome is localized at the infection site in the lung and consequently may fully exhibit its in vivo activity. The efficacy of AmBisome is superior to that of Fungizone against pulmonary aspergillosis.
Collapse
Affiliation(s)
- Koji Takemoto
- Discovery Research Laboratories II, Sumitomo Pharmaceuticals Research Division, Konohana-ku, Osaka, Japan.
| | | | | | | | | | | |
Collapse
|
31
|
Mukherjee PK, Sheehan DJ, Hitchcock CA, Ghannoum MA. Combination treatment of invasive fungal infections. Clin Microbiol Rev 2005; 18:163-94. [PMID: 15653825 PMCID: PMC544182 DOI: 10.1128/cmr.18.1.163-194.2005] [Citation(s) in RCA: 209] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The persistence of high morbidity and mortality from systemic fungal infections despite the availability of novel antifungals points to the need for effective treatment strategies. Treatment of invasive fungal infections is often hampered by drug toxicity, tolerability, and specificity issues, and added complications often arise due to the lack of diagnostic tests and to treatment complexities. Combination therapy has been suggested as a possible approach to improve treatment outcome. In this article, we undertake a historical review of studies of combination therapy and also focus on recent studies involving newly approved antifungal agents. The limitations surrounding antifungal combinations include nonuniform interpretation criteria, inability to predict the likelihood of clinical success, strain variability, and variations in pharmacodynamic/pharmacokinetic properties of antifungals used in combination. The issue of antagonism between polyenes and azoles is beginning to be addressed, but data regarding other drug combinations are not adequate for us to draw definite conclusions. However, recent data have identified potentially useful combinations. Standardization of assay methods and adoption of common interpretive criteria are essential to avoid discrepancies between different in vitro studies. Larger clinical trials are needed to assess whether combination therapy improves survival and treatment outcome in the most seriously debilitated patients afflicted with life-threatening fungal infections.
Collapse
Affiliation(s)
- Pranab K Mukherjee
- Center for Medical Mycology, Department of Dermatology, Case Western Reserve University and University Hospitals of Cleveland, 11100 Euclid Ave., LKS-5028, Cleveland, OH 44106-5028, USA
| | | | | | | |
Collapse
|
32
|
Bellocchio S, Gaziano R, Bozza S, Rossi G, Montagnoli C, Perruccio K, Calvitti M, Pitzurra L, Romani L. Liposomal amphotericin B activates antifungal resistance with reduced toxicity by diverting Toll-like receptor signalling from TLR-2 to TLR-4. J Antimicrob Chemother 2005; 55:214-22. [PMID: 15649994 DOI: 10.1093/jac/dkh542] [Citation(s) in RCA: 98] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
OBJECTIVES Neutrophils play a crucial role in the control of the Aspergillus fumigatus infection and act in concert with antifungal drugs. This study was undertaken to obtain insights into the possible involvement of Toll-like receptors (TLRs) in the interaction of liposomal amphotericin B (L-AmB; AmBisome) with neutrophils in response to A. fumigatus. METHODS For generation of bone marrow-transplanted mice, irradiated C57BL6 mice were infused with T cell-depleted allogeneic donor cells. For infection, mice were injected intranasally with Aspergillus fumigatus conidia and treated with L-Amb and deoxycholate amphotericin B prophylactically or therapeutically. For TLR-dependent antifungal functions, murine neutrophils were preincubated with antifungals or TLR ligands before the addition of Aspergillus conidia. RESULTS The results show that: (a) neutrophil activation by Aspergillus occurs through TLR signalling pathways differently affecting the oxidative and non-oxidative mechanisms of the killing machinery; (b) by diverting signalling from TLR-2 to TLR-4, liposomes of AmBisome activate neutrophils to an antifungal state while attenuating the pro-inflammatory effects of deoxycholate amphotericin B; (c) this translates in vivo to the optimization of the AmBisome therapeutic efficacy in mice with aspergillosis. CONCLUSIONS These results provide a putative molecular basis for the reduced infusion-related toxicity of AmBisome and suggest that TLR manipulation in vivo is amenable to the induction of optimal microbicidal activity in the absence of inflammatory cytotoxicity to host cells.
Collapse
Affiliation(s)
- Silvia Bellocchio
- Microbiology Sections, Department of Experimental Medicine and Biochemical Sciences, University of Perugia, Perugia, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Gil-Lamaignere C, Winn RM, Simitsopoulou M, Maloukou A, Walsh TJ, Roilides E. Inteferon gamma and granulocyte–macrophage colony-stimulating factor augment the antifungal activity of human polymorphonuclear leukocytes againstScedosporiumspp.: comparison withAspergillusspp. Med Mycol 2005; 43:253-60. [PMID: 16010852 DOI: 10.1080/13693780412331271072] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Abstract
While Aspergillus spp. have been the most frequent filamentous fungi causing infections in immunocompromised patients, Scedosporium spp. are emerging as life-threatening pathogens. We studied the effects of interferon gamma (IFN-gamma) and granulocyte-macrophage colony-stimulating factor (GM-CSF) alone or combined on the antifungal activities of human polymorphonuclear leukocytes (PMN) against Scedosporium apiospermum and Scedosporium prolificans. We paralleled these activities to those against Aspergillus fumigatus and Aspergillus flavus. Incubation of PMN with IFN-gamma and GM-CSF for 22 h enhanced PMN-induced hyphal damage of both Aspergillus spp. and S. prolificans (p < 0.05) but not of S. apiospermum. However, hyphae of S. apiospermum were damaged significantly more after incubation with PMN that had been treated with IFN-gamma and GM-CSF for 2 h. In addition, incubation of PMN with GM-CSF for 2 h enhanced PMN oxidative burst measured as superoxide anion (O2-) production in response to nonopsonized hyphae of A. flavus and Scedosporium spp. (p < 0.05). In contrast, after 2 h, IFN-gamma and GM-CSF alone did not enhance PMN O2- in response to opsonized hyphae of A. flavus and Scedosporium spp.; however, the combination of IFN-gamma and GM-CSF showed significant enhancement against these species. Thus, IFN-gamma and GM-CSF, particularly in combination, demonstrate a species- and time-dependent augmentation of PMN responses to Scedosporium spp.
Collapse
Affiliation(s)
- Cristina Gil-Lamaignere
- Third Department of Pediatrics, Aristotle University of Thessaloniki, Hippokration Hospital, Thessaloniki, Greece
| | | | | | | | | | | |
Collapse
|
34
|
Gaziano R, Bozza S, Bellocchio S, Perruccio K, Montagnoli C, Pitzurra L, Salvatori G, De Santis R, Carminati P, Mantovani A, Romani L. Anti-Aspergillus fumigatus efficacy of pentraxin 3 alone and in combination with antifungals. Antimicrob Agents Chemother 2004; 48:4414-21. [PMID: 15504871 PMCID: PMC525434 DOI: 10.1128/aac.48.11.4414-4421.2004] [Citation(s) in RCA: 114] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The collectin pentraxin 3 (PTX3) is an essential component of host resistance to pulmonary aspergillosis. Here we examined the protective effects of administration of PTX3 alone or together with deoxycholate amphotericin B (Fungizone) or liposomal amphotericin B (AmBisome) against invasive aspergillosis in a murine model of allogeneic bone marrow transplantation. PTX3, alone or in combination with the polyenes, was given intranasally or parenterally either before, in concomitance with, or after the intranasal infection with Aspergillus fumigatus conidia. Mice were monitored for resistance to infection and parameters of innate and adaptive T-helper immunity. The results showed the following: (i) complete resistance to infection and reinfection was observed in mice treated with PTX3 alone; (ii) the protective effect of PTX3 was similar or superior to that observed with liposomal amphotericin B or deoxycholate amphotericin B, respectively; (iii) protection was associated with accelerated recovery of lung phagocytic cells and T-helper-1 lymphocytes and concomitant decrease of inflammatory pathology; and (iv) PTX3 potentiated the therapeutic efficacy of suboptimal doses of either antimycotic drug. Together, these data suggest the potential therapeutic use of PTX3 either alone or as an adjunctive therapy in A. fumigatus infections.
Collapse
Affiliation(s)
- Roberta Gaziano
- Department of Experimental Medicine and Biochemical Sciences, Microbiology Section, University of Perugia, Perugia, Sigma-Tau Industrie Farmaceutiche Riunite S.P.A, Pomezia, Rome, Department of Immunology and Cell Biology, Mario Negri Institute, Milan, Italy
| | - Silvia Bozza
- Department of Experimental Medicine and Biochemical Sciences, Microbiology Section, University of Perugia, Perugia, Sigma-Tau Industrie Farmaceutiche Riunite S.P.A, Pomezia, Rome, Department of Immunology and Cell Biology, Mario Negri Institute, Milan, Italy
| | - Silvia Bellocchio
- Department of Experimental Medicine and Biochemical Sciences, Microbiology Section, University of Perugia, Perugia, Sigma-Tau Industrie Farmaceutiche Riunite S.P.A, Pomezia, Rome, Department of Immunology and Cell Biology, Mario Negri Institute, Milan, Italy
| | - Katia Perruccio
- Department of Experimental Medicine and Biochemical Sciences, Microbiology Section, University of Perugia, Perugia, Sigma-Tau Industrie Farmaceutiche Riunite S.P.A, Pomezia, Rome, Department of Immunology and Cell Biology, Mario Negri Institute, Milan, Italy
| | - Claudia Montagnoli
- Department of Experimental Medicine and Biochemical Sciences, Microbiology Section, University of Perugia, Perugia, Sigma-Tau Industrie Farmaceutiche Riunite S.P.A, Pomezia, Rome, Department of Immunology and Cell Biology, Mario Negri Institute, Milan, Italy
| | - Lucia Pitzurra
- Department of Experimental Medicine and Biochemical Sciences, Microbiology Section, University of Perugia, Perugia, Sigma-Tau Industrie Farmaceutiche Riunite S.P.A, Pomezia, Rome, Department of Immunology and Cell Biology, Mario Negri Institute, Milan, Italy
| | - Giovanni Salvatori
- Department of Experimental Medicine and Biochemical Sciences, Microbiology Section, University of Perugia, Perugia, Sigma-Tau Industrie Farmaceutiche Riunite S.P.A, Pomezia, Rome, Department of Immunology and Cell Biology, Mario Negri Institute, Milan, Italy
| | - Rita De Santis
- Department of Experimental Medicine and Biochemical Sciences, Microbiology Section, University of Perugia, Perugia, Sigma-Tau Industrie Farmaceutiche Riunite S.P.A, Pomezia, Rome, Department of Immunology and Cell Biology, Mario Negri Institute, Milan, Italy
| | - Paolo Carminati
- Department of Experimental Medicine and Biochemical Sciences, Microbiology Section, University of Perugia, Perugia, Sigma-Tau Industrie Farmaceutiche Riunite S.P.A, Pomezia, Rome, Department of Immunology and Cell Biology, Mario Negri Institute, Milan, Italy
| | - Alberto Mantovani
- Department of Experimental Medicine and Biochemical Sciences, Microbiology Section, University of Perugia, Perugia, Sigma-Tau Industrie Farmaceutiche Riunite S.P.A, Pomezia, Rome, Department of Immunology and Cell Biology, Mario Negri Institute, Milan, Italy
| | - Luigina Romani
- Department of Experimental Medicine and Biochemical Sciences, Microbiology Section, University of Perugia, Perugia, Sigma-Tau Industrie Farmaceutiche Riunite S.P.A, Pomezia, Rome, Department of Immunology and Cell Biology, Mario Negri Institute, Milan, Italy
- Corresponding author. Mailing address: Department of Experimental Medicine and Biochemical Sciences—Microbiology Section, University of Perugia, Via del Giochetto, 06122 Perugia, Italy. Phone and fax: 039-075-585-7411. E-mail:
| |
Collapse
|
35
|
Abstract
Invasive fungal infections pose major management problems for clinicians caring for hematopoietic cell transplant patients. Two major fungal genera, Candida and Aspergillus, account for most fungal infections. Rates of systemic Candida infection range from 15% to 25%, mostly in the pre-engraftment period. Prophylaxis by fluconazole has dramatically reduced the frequency of early Candida infections. Caspofungin has recently been shown to offer an excellent alternative to amphotericin B (with less toxicity) or fluconazole (with a broader spectrum) for therapy of systemic Candida infections. Aspergillus infections occur in 15% to 20% of allogeneic hematopoietic cell transplant patients, most frequently in the post-engraftment period; they are associated with a severe diminution of cell-mediated immune responses by graft-versus-host disease and prolonged corticosteroid use. Voriconazole, a recently introduced broad-spectrum azole, has excellent activity against Aspergillus and is generally well tolerated. Voriconazole currently offers the best prospect for success and tolerance as a first-line treatment for aspergillosis. Second-line therapies include lipid formulations of amphotericin B, caspofungin, or intravenous itraconazole. Unfortunately, early initiation of therapy for aspergillosis is frequently not possible because of inaccurate diagnostics. One new diagnostic, the galactomannan assay, has recently been approved, and others are in development; these offer promise for earlier diagnosis without the need for invasive procedures. It is hoped that these new therapies and new diagnostics will usher in a new era of antifungal therapy.
Collapse
Affiliation(s)
- John R Wingard
- Blood and Marrow Transplant Program, Division of Hematology/Oncology, University of Florida Shands Cancer Center, Gainesville, USA.
| | | |
Collapse
|
36
|
Choi JH, Brummer E, Stevens DA. Combined action of micafungin, a new echinocandin, and human phagocytes for antifungal activity against Aspergillus fumigatus. Microbes Infect 2004; 6:383-9. [PMID: 15050966 DOI: 10.1016/j.micinf.2003.12.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2003] [Accepted: 12/17/2003] [Indexed: 10/26/2022]
Abstract
Micafungin, a new echinocandin, inhibits fungal cell wall beta-glucan synthesis. We postulated micafungin and host phagocytic cells could act together in damaging fungi. Using the metabolic XTT assay, micafungin alone (0.01 and 0.10 microg/ml) inhibited Aspergillus fumigatus germlings by 48% and 61%, respectively. Polymorphonuclear neutrophils (PMNs) inhibited germlings by 53%. Micafungin at 0.01 or 0.10 microg/ml and PMNs resulted in additive inhibition, 82% and 99%, respectively. Monocyte-derived macrophage (MDM) monolayers inhibited germling growth by 66%; micafungin (0.01 or 0.10 microg/ml) alone inhibited by 32% and 42%, respectively. MDMs and micafungin (0.01 or 0.10 microg/ml) caused an additive inhibition of growth, 85% and 95%, respectively. Hyphae were generated by incubation of conidia for 24 h with or without micafungin. PMNs alone, added to hyphae, inhibited growth by 19% in the subsequent 20 h. Hyphae generated in the presence of micafungin (0.10 microg/ml) and subsequently cultured with micafungin for 24 h inhibited growth by 64%. PMNs plus micafungin resulted in 82% inhibition. Monocytes alone inhibited hyphal growth by only 5%. Hyphae produced in the presence of micafungin (0.01 microg/ml) and incubated again with micafungin for 24 h inhibited growth by 47%; combination with monocytes resulted in 62% inhibition. These data indicate that micafungin inhibits growth of tissue forms of A. fumigatus, and phagocytes and micafungin together have an additive effect. These findings support the thesis that the greater efficacy of micafungin in vivo compared with in vitro could be due to combined effect of phagocytic cells and micafungin.
Collapse
Affiliation(s)
- Jung-Hyun Choi
- Department of Medicine, Division of Infectious Diseases, Santa Clara Valley Medical Center, San Jose, CA 95128-2699, USA
| | | | | |
Collapse
|
37
|
|