1
|
Padilla Suarez EG, Siciliano A, Spampinato M, Maione A, Guida M, Libralato G, Galdiero E. Ecotoxicity and Mutagenicity Assessment of Novel Antifungal Agents VT-1161 and T-2307. Molecules 2024; 29:4739. [PMID: 39407667 PMCID: PMC11477875 DOI: 10.3390/molecules29194739] [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: 09/10/2024] [Revised: 09/30/2024] [Accepted: 10/02/2024] [Indexed: 10/20/2024] Open
Abstract
Antifungal substances are essential for managing fungal infections in humans, animals, and plants, and their usage has significantly increased due to the global rise in fungal infections. However, the extensive application of antifungal agents in pharmaceuticals, personal care products, and agriculture has led to their widespread environmental dissemination through various pathways, such as excretion, improper disposal, and agricultural runoff. Despite advances in wastewater treatment, many antifungal compounds persist in the environment, affecting non-target organisms and contributing to resistance development. This study investigates the environmental impact of two novel antifungal agents, VT-1161 and T-2307, recently introduced as alternatives for treating resistant Candida spp. We assessed their ecotoxicity and mutagenicity using multiple bioassays: immobilization of Daphnia magna, growth inhibition of Raphidocelis subcapitata, luminescence inhibition of Aliivibrio fischeri, and mutagenicity on Salmonella typhimurium strain TA100. Results indicate that both VT-1161 and T-2307 exhibit lower toxicity compared to existing antifungal compounds, with effective concentrations (EC50) causing 50% response ranging from 14.34 to 27.92 mg L-1. Furthermore, both agents were classified as less hazardous based on the Globally Harmonized System of Classification and Labeling of Chemicals. Despite these favorable results, further research is needed to understand their environmental behavior, interactions, and potential resistance development among non-target species. Our findings highlight the importance of comprehensive environmental risk assessments to ensure the sustainable use of new antifungal agents.
Collapse
Affiliation(s)
- Edith Guadalupe Padilla Suarez
- Department of Biology, University of Naples Federico II, Complesso Universitario Monte Sant’Angelo, Via Cintia 4, 80126 Naples, Italy; (E.G.P.S.); (M.S.); (A.M.); (M.G.); (G.L.); (E.G.)
| | - Antonietta Siciliano
- Department of Biology, University of Naples Federico II, Complesso Universitario Monte Sant’Angelo, Via Cintia 4, 80126 Naples, Italy; (E.G.P.S.); (M.S.); (A.M.); (M.G.); (G.L.); (E.G.)
| | - Marisa Spampinato
- Department of Biology, University of Naples Federico II, Complesso Universitario Monte Sant’Angelo, Via Cintia 4, 80126 Naples, Italy; (E.G.P.S.); (M.S.); (A.M.); (M.G.); (G.L.); (E.G.)
- NBFC—National Biodiversity Future Center, 90133 Palermo, Italy
| | - Angela Maione
- Department of Biology, University of Naples Federico II, Complesso Universitario Monte Sant’Angelo, Via Cintia 4, 80126 Naples, Italy; (E.G.P.S.); (M.S.); (A.M.); (M.G.); (G.L.); (E.G.)
| | - Marco Guida
- Department of Biology, University of Naples Federico II, Complesso Universitario Monte Sant’Angelo, Via Cintia 4, 80126 Naples, Italy; (E.G.P.S.); (M.S.); (A.M.); (M.G.); (G.L.); (E.G.)
- NBFC—National Biodiversity Future Center, 90133 Palermo, Italy
| | - Giovanni Libralato
- Department of Biology, University of Naples Federico II, Complesso Universitario Monte Sant’Angelo, Via Cintia 4, 80126 Naples, Italy; (E.G.P.S.); (M.S.); (A.M.); (M.G.); (G.L.); (E.G.)
| | - Emilia Galdiero
- Department of Biology, University of Naples Federico II, Complesso Universitario Monte Sant’Angelo, Via Cintia 4, 80126 Naples, Italy; (E.G.P.S.); (M.S.); (A.M.); (M.G.); (G.L.); (E.G.)
| |
Collapse
|
2
|
Morrissey CO, Kim HY, Garnham K, Dao A, Chakrabarti A, Perfect JR, Alastruey-Izquierdo A, Harrison TS, Bongomin F, Galas M, Siswanto S, Dagne DA, Roitberg F, Gigante V, Sati H, Alffenaar JW, Beardsley J. Mucorales: A systematic review to inform the World Health Organization priority list of fungal pathogens. Med Mycol 2024; 62:myad130. [PMID: 38935901 PMCID: PMC11210621 DOI: 10.1093/mmy/myad130] [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: 09/17/2023] [Revised: 11/18/2023] [Accepted: 12/11/2023] [Indexed: 06/29/2024] Open
Abstract
The World Health Organization, in response to the growing burden of fungal disease, established a process to develop a fungal priority pathogens list (FPPL). This systematic review aimed to evaluate the epidemiology and impact of invasive fungal disease due to Mucorales. PubMed and Web of Science were searched to identify studies published between January 1, 2011 and February 23, 2021. Studies reporting on mortality, inpatient care, complications and sequelae, antifungal susceptibility, risk factors, preventability, annual incidence, global distribution, and emergence during the study time frames were selected. Overall, 24 studies were included. Mortality rates of up to 80% were reported. Antifungal susceptibility varied across agents and species, with the minimum inhibitory concentrations lowest for amphotericin B and posaconazole. Diabetes mellitus was a common risk factor, detected in 65%-85% of patients with mucormycosis, particularly in those with rhino-orbital disease (86.9%). Break-through infection was detected in 13.6%-100% on azole or echinocandin antifungal prophylaxis. The reported prevalence rates were variable, with some studies reporting stable rates in the USA of 0.094-0.117/10 000 discharges between 2011 and 2014, whereas others reported an increase in Iran from 16.8% to 24% between 2011 and 2015. Carefully designed global surveillance studies, linking laboratory and clinical data, are required to develop clinical breakpoints to guide antifungal therapy and determine accurate estimates of complications and sequelae, annual incidence, trends, and global distribution. These data will provide robust estimates of disease burden to refine interventions and better inform future FPPL.
Collapse
Affiliation(s)
- C Orla Morrissey
- Department of Infectious Diseases, Alfred Health and Monash University, Melbourne, Victoria, Australia
| | - Hannah Yejin Kim
- Infectious Diseases Institute (Sydney ID), The University of Sydney, Camperdown, New South Wales, Australia
- Faculty of Medicine and Health, School of Pharmacy, The University of Sydney, Camperdown, New South Wales, Australia
- Department of Pharmacy, Westmead Hospital, Westmead, New South Wales, Australia
| | - Katherine Garnham
- Department of Infectious Diseases and Microbiology, Gold Coast University Hospital, Southport, Queensland, Australia
| | - Aiken Dao
- Infectious Diseases Institute (Sydney ID), The University of Sydney, Camperdown, New South Wales, Australia
- Department of Infectious Diseases, Sydney Medical School, Faculty of Medicine and Health, University of Sydney, Camperdown, New South Wales, Australia
- Orthopaedic Research and Biotechnology Unit, Children’s Hospital at Westmead, Westmead, New South Wales, Australia
| | | | - John R Perfect
- Division of Infectious Diseases and International Health, Duke University School of Medicine, Durham, North Carolina, USA
| | - Ana Alastruey-Izquierdo
- Mycology Reference Laboratory, National Centre for Microbiology, Instituto de Salud Carlos III, Majadahonda, Madrid, Spain
| | - Thomas S Harrison
- Institute for Infection and Immunity, and Clinical Academic Group in Infection and Immunity, St. George’s, University of London, and St. George's University Hospitals NHS Foundation Trust, London, United Kingdom
- MRC Centre for Medical Mycology, University of Exeter, Exeter, United Kingdom
| | - Felix Bongomin
- Department of Medical Microbiology and Immunology, Faculty of Medicine, Gulu University, Gulu, Uganda
| | - Marcelo Galas
- Antimicrobial Resistance Special Program, Communicable Diseases and Environmental Determinants of Health, Pan American Health Organization, Washington, District of Columbia, USA
| | - Siswanto Siswanto
- World Health Organization, South-East Asia Region Office, New Delhi, India
| | - Daniel Argaw Dagne
- Department of Control of Neglected Tropical Diseases, World Health Organization, Geneva, Switzerland
| | - Felipe Roitberg
- Department of Noncommunicable Diseases, World Health Organization, Geneva, Switzerland
| | - Valeria Gigante
- AMR Division, World Health Organization, Geneva, Switzerland
| | - Hatim Sati
- AMR Division, World Health Organization, Geneva, Switzerland
| | - Jan-Willem Alffenaar
- Infectious Diseases Institute (Sydney ID), The University of Sydney, Camperdown, New South Wales, Australia
- Faculty of Medicine and Health, School of Pharmacy, The University of Sydney, Camperdown, New South Wales, Australia
- Department of Pharmacy, Westmead Hospital, Westmead, New South Wales, Australia
| | - Justin Beardsley
- Infectious Diseases Institute (Sydney ID), The University of Sydney, Camperdown, New South Wales, Australia
- Department of Pharmacy, Westmead Hospital, Westmead, New South Wales, Australia
- Westmead Institute for Medical Research, Westmead, New South Wales, Australia
| |
Collapse
|
3
|
Little JS, Kampouri E, Friedman DZ, McCarty T, Thompson GR, Kontoyiannis DP, Vazquez J, Baddley JW, Hammond SP. The Burden of Invasive Fungal Disease Following Chimeric Antigen Receptor T-Cell Therapy and Strategies for Prevention. Open Forum Infect Dis 2024; 11:ofae133. [PMID: 38887472 PMCID: PMC11181190 DOI: 10.1093/ofid/ofae133] [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: 12/01/2023] [Accepted: 03/05/2024] [Indexed: 06/20/2024] Open
Abstract
Chimeric antigen receptor (CAR) T-cell therapy is a novel immunotherapy approved for the treatment of hematologic malignancies. This therapy leads to a variety of immunologic deficits that could place patients at risk for invasive fungal disease (IFD). Studies assessing IFD in this setting are limited by inconsistent definitions and heterogeneity in prophylaxis use, although the incidence of IFD after CAR T-cell therapy, particularly for lymphoma and myeloma, appears to be low. This review evaluates the incidence of IFD after CAR T-cell therapy, and discusses optimal approaches to prevention, highlighting areas that require further study as well as future applications of cellular therapy that may impact IFD risk. As the use of CAR T-cell therapy continues to expand for hematologic malignancies, solid tumors, and most recently to include non-oncologic diseases, understanding the risk for IFD in this uniquely immunosuppressed population is imperative to prevent morbidity and mortality.
Collapse
Affiliation(s)
- Jessica S Little
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
- Division of Infectious Diseases, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Eleftheria Kampouri
- Infectious Diseases Service, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Daniel Z Friedman
- Section of Infectious Diseases and Global Health, The University of Chicago, Chicago, Illinois, USA
| | - Todd McCarty
- Division of Infectious Diseases, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - George R Thompson
- Division of Infectious Diseases, University of California-Davis, Sacramento, California, USA
| | - Dimitrios P Kontoyiannis
- Department of Infectious Diseases, Infection Control and Employee Health, The University of Texas, M.D. Anderson Cancer Center, Houston, Texas, USA
| | - Jose Vazquez
- Division of Infectious Diseases, Medical College of Georgia/Augusta University, Augusta, Georgia, USA
| | - John W Baddley
- Division of Infectious Diseases, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Sarah P Hammond
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Medical Oncology, Massachusetts General Hospital Cancer Center, Boston, Massachusetts, USA
| |
Collapse
|
4
|
Marchand G, Wingert L, Viegas C, Caetano L, Viegas S, Twaruzek M, Lacombe N, Lanoie D, Valois I, Gouin F, Soszczyńska E, Kosicki R, Dias M, Debia M. Assessment of waste workers occupational risk to microbial agents and cytotoxic effects of mixed contaminants present in the air of waste truck cabin and ventilation filters. JOURNAL OF THE AIR & WASTE MANAGEMENT ASSOCIATION (1995) 2024; 74:145-162. [PMID: 38166349 DOI: 10.1080/10962247.2023.2299424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 12/20/2023] [Indexed: 01/04/2024]
Abstract
Workers in the waste-processing industry are potentially exposed to high concentrations of biological contaminants, leading to respiratory and digestive problems and skin irritations. However, few data on the exposure of waste collection truck (WCT) drivers are available. The goal was to document the microbial risk of the waste collection truck (WCT) workers while in the vehicle cab. Long-period sampling using the truck air filters (CAF) and short time ambient air sampling in the cab were used. The potential release of microbial particles from CAFs was also investigated since it could contribute to the microbial load of the cabin air. A combination of analytical methods also helped assess the complex mixture of the biological agents. Aspergillus sections Fumigati and Flavi, E. coli, Enterobacter spp. and Legionella spp. were detected in the CAF of trucks collecting three types of waste. The highest levels of bacteria and fungi were found in the CAF from organic WCT. The highest endotoxin concentrations in CAF were 300 EU/cm2. Most of the CAF showed cytotoxic effects on both lung cells and hepatocytes. Only one mycotoxin was detected in a CAF. The maximal concentrations in the ambient WCT air varied according to the type of waste collected. The highest proportion (84%) of the air samples without cytotoxic effects on the lungs cells was for the recyclable material WCTs. The results revealed the potential microbial risk to workers from a complex mixture of bio-contaminants in the cabs of vehicles collecting all types of waste. The sustained cytotoxic effect indicates the potential adverse health-related impact of mixed contaminants (biological and non-biological) for the workers. Overall, this study highlights the benefits of using complementary sampling strategy and combined analytical methods for a the assessment of the microbial risk in work environments and the need to implement protective measures for the workers.Implications: Exposure to microbial agents is a well-known occupational hazard in the waste management sector. No previous study had evaluated the cytotoxicity of ambient air and ventilation filters to document worker exposure to a combination of contaminants during waste collection. This research confirms the usefulness of ventilation filters for long-term characterization of exposure to infectious agents, azole-resistant fungi, coliform bacteria and mycotoxin. Overall, this study highlights the importance of using several sampling and analysis methods for a comprehensive assessment of microbial risk in work environments, as well as the need to implement appropriate protective measures for collection workers.
Collapse
Affiliation(s)
- Genevieve Marchand
- Department of Environmental and Occupational Health, School of Public Health, Université de Montréal, Montreal, Canada
- Institut de recherche Robert-Sauvé en santé et en sécurité du travail, Montreal, Canada
| | - Loïc Wingert
- Department of Environmental and Occupational Health, School of Public Health, Université de Montréal, Montreal, Canada
- Institut de recherche Robert-Sauvé en santé et en sécurité du travail, Montreal, Canada
| | - Carla Viegas
- H&TRC - Health & Technology Research Center, ESTeSL - Escola Superior de Tecnologia e Saúde, In-stituto Politécnico de Lisboa, Lisboa, Portugal
- NOVA National School of Public Health, Public Health Research Centre, Comprehensive Health Research Center, CHRC, NOVA University Lisbon, Lisbon, Portugal
| | - Liliana Caetano
- H&TRC - Health & Technology Research Center, ESTeSL - Escola Superior de Tecnologia e Saúde, In-stituto Politécnico de Lisboa, Lisboa, Portugal
- Research Institute for Medicines (iMed. ULisboa), Faculty of Pharmacy, University of Lisbon, Lisbon, Portugal
| | - Susana Viegas
- NOVA National School of Public Health, Public Health Research Centre, Comprehensive Health Research Center, CHRC, NOVA University Lisbon, Lisbon, Portugal
| | - Magdalena Twaruzek
- Faculty of Biological Sciences, Department of Physiology and Toxicology, Kazimierz Wielki University, Bydgoszcz, Poland
| | - Nancy Lacombe
- Institut de recherche Robert-Sauvé en santé et en sécurité du travail, Montreal, Canada
| | - Delphine Lanoie
- Institut de recherche Robert-Sauvé en santé et en sécurité du travail, Montreal, Canada
| | - Isabelle Valois
- Department of Environmental and Occupational Health, School of Public Health, Université de Montréal, Montreal, Canada
| | - Francois Gouin
- Institut de recherche Robert-Sauvé en santé et en sécurité du travail, Montreal, Canada
| | - Ewelina Soszczyńska
- Faculty of Biological Sciences, Department of Physiology and Toxicology, Kazimierz Wielki University, Bydgoszcz, Poland
| | - Robert Kosicki
- Faculty of Biological Sciences, Department of Physiology and Toxicology, Kazimierz Wielki University, Bydgoszcz, Poland
| | - Marta Dias
- H&TRC - Health & Technology Research Center, ESTeSL - Escola Superior de Tecnologia e Saúde, In-stituto Politécnico de Lisboa, Lisboa, Portugal
- Research Institute for Medicines (iMed. ULisboa), Faculty of Pharmacy, University of Lisbon, Lisbon, Portugal
| | - Maximilien Debia
- Department of Environmental and Occupational Health, School of Public Health, Université de Montréal, Montreal, Canada
| |
Collapse
|
5
|
Little JS, Duléry R, Shapiro RM, Aleissa MM, Prockop SE, Koreth J, Ritz J, Antin JH, Cutler C, Nikiforow S, Romee R, Issa NC, Ho VT, Baden LR, Soiffer RJ, Gooptu M. Opportunistic Infections in Patients Receiving Post-Transplantation Cyclophosphamide: Impact of Haploidentical versus Unrelated Donor Allograft. Transplant Cell Ther 2024; 30:233.e1-233.e14. [PMID: 37984797 DOI: 10.1016/j.jtct.2023.11.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 11/13/2023] [Accepted: 11/14/2023] [Indexed: 11/22/2023]
Abstract
Post-transplantation cyclophosphamide (PTCy) is an effective strategy for graft-versus-host disease (GVHD) prophylaxis and is the standard of care for haploidentical hematopoietic cell transplantation (HCT). It is increasingly used for matched and mismatched unrelated donor (MUD/MMUD) HCT, but infections remain a concern. The objective of this study was to evaluate the characteristics and risk factors for infections in haploidentical and unrelated donor HCT recipients treated with PTCy-based GVHD prophylaxis. This single-center retrospective study examined 354 consecutive adults undergoing HCT with PTCy-based GVHD prophylaxis (161 MUD/MMUD; 193 haploidentical) between 2015 and 2022. Opportunistic infections (OIs), including cytomegalovirus (CMV), adenovirus (AdV), Epstein-Barr virus (EBV), and invasive fungal disease (IFD), were assessed from day 0 through day +365. The 1-year cumulative incidence functions of OIs and nonrelapse mortality (NRM) were calculated using dates of relapse and repeat HCT as competing risks. Secondary analysis evaluated risk factors for OIs and NRM using univariate and multivariable Cox regression models. Haploidentical HCT recipients had an increased risk of OIs compared to unrelated donor allograft recipients (39% for haploidentical versus 25% for MUD/MMUD; hazard ratio [HR], 1.70; 95% confidence interval [CI], 1.16 to 2.49; P = .006). On multivariable analysis, haploidentical donor (HR, 1.50; 95% CI, 1.01 to 2.23; P = .046), prior HCT (HR, 1.99; 95% CI, 1.29 to 3.09; P = .002), and diagnosis of aGVHD (HR, 1.47; 95% CI, 1.02 to 2.14; P = .041) were associated with increased risk of OIs. NRM within the first year was not significantly different between the 2 cohorts (HR, 1.11; 95% CI, .64 to 1.93; P = .70). Overall, haploidentical donor was a significant risk factor for OIs in patients receiving PTCy, although 1-year NRM was not different between haploidentical HCT and MUD/MMUD HCT recipients. CMV and AdV infections were significantly increased among haploidentical HCT recipients, whereas the incidences of EBV infection and IFD were similar in the 2 cohorts. Our findings may have implications for infection monitoring and prophylaxis in the setting of PTCy, particularly in haploidentical HCT recipients.
Collapse
Affiliation(s)
- Jessica S Little
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, Massachusetts; Harvard Medical School, Boston, Massachusetts; Stem Cell Transplant and Cellular Therapy, Dana-Farber Cancer Institute, Boston, Massachusetts.
| | - Rémy Duléry
- Stem Cell Transplant and Cellular Therapy, Dana-Farber Cancer Institute, Boston, Massachusetts; Sorbonne University, Department of Clinical Hematology and Cellular Therapy, Saint-Antoine Hospital, Assistance Publique-Hôpitaux de Paris, Inserm UMRs 938, Centre de recherche Saint-Antoine, Paris, France
| | - Roman M Shapiro
- Harvard Medical School, Boston, Massachusetts; Stem Cell Transplant and Cellular Therapy, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Muneerah M Aleissa
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, Massachusetts; Harvard Medical School, Boston, Massachusetts; Department of Pharmacy Practice, College of Pharmacy, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Susan E Prockop
- Harvard Medical School, Boston, Massachusetts; Hematopoietic Stem Cell Transplant Program, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, Massachusetts
| | - John Koreth
- Harvard Medical School, Boston, Massachusetts; Stem Cell Transplant and Cellular Therapy, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Jerome Ritz
- Harvard Medical School, Boston, Massachusetts; Stem Cell Transplant and Cellular Therapy, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Joseph H Antin
- Harvard Medical School, Boston, Massachusetts; Stem Cell Transplant and Cellular Therapy, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Corey Cutler
- Harvard Medical School, Boston, Massachusetts; Stem Cell Transplant and Cellular Therapy, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Sarah Nikiforow
- Harvard Medical School, Boston, Massachusetts; Stem Cell Transplant and Cellular Therapy, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Rizwan Romee
- Harvard Medical School, Boston, Massachusetts; Stem Cell Transplant and Cellular Therapy, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Nicolas C Issa
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, Massachusetts; Harvard Medical School, Boston, Massachusetts; Stem Cell Transplant and Cellular Therapy, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Vincent T Ho
- Harvard Medical School, Boston, Massachusetts; Stem Cell Transplant and Cellular Therapy, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Lindsey R Baden
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, Massachusetts; Harvard Medical School, Boston, Massachusetts; Stem Cell Transplant and Cellular Therapy, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Robert J Soiffer
- Harvard Medical School, Boston, Massachusetts; Stem Cell Transplant and Cellular Therapy, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Mahasweta Gooptu
- Harvard Medical School, Boston, Massachusetts; Stem Cell Transplant and Cellular Therapy, Dana-Farber Cancer Institute, Boston, Massachusetts
| |
Collapse
|
6
|
Walker J, Edwards WS, Hall NM, Pappas PG. Challenges in management of invasive fungal infections in stem cell transplant. Transpl Infect Dis 2023; 25 Suppl 1:e14175. [PMID: 37864814 DOI: 10.1111/tid.14175] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 10/02/2023] [Accepted: 10/03/2023] [Indexed: 10/23/2023]
Abstract
Invasive fungal infections cause significant morbidity and mortality in hematopoietic stem cell transplant recipients. In order to minimize these infections, prophylaxis has become routine, although the agents used have changed over time. This presents new challenges as we consider an approach to breakthrough infections and recognize the epidemiologic shift toward isolates with higher rates of drug resistance. This review outlines the management of the most common pathogens (Candida, Aspergillus, Mucorales) as well as rarer pathogens that have higher rates of resistance (Trichosporon, Fusarium, Scedosporium, and Lomentospora). We discuss potential approaches to proven or possible breakthrough infections with yeast and pulmonary mold disease. Finally, we outline the role for combination therapy and newer antifungals, acknowledging current knowledge gaps and areas for future exploration.
Collapse
Affiliation(s)
- Jeremey Walker
- Division of Infectious Diseases, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - W Seth Edwards
- Department of Pharmacy, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Nicole M Hall
- Department of Pharmacy, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Peter G Pappas
- Division of Infectious Diseases, University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
7
|
Puerta-Alcalde P, Monzó-Gallo P, Aguilar-Guisado M, Ramos JC, Laporte-Amargós J, Machado M, Martin-Davila P, Franch-Sarto M, Sánchez-Romero I, Badiola J, Gómez L, Ruiz-Camps I, Yáñez L, Vázquez L, Chumbita M, Marco F, Soriano A, González P, Fernández-Cruz A, Batlle M, Fortún J, Guinea J, Gudiol C, García J, Ruiz Pérez de Pipaón M, Alastruey-Izquierdo A, Garcia-Vidal C. Breakthrough invasive fungal infection among patients with haematologic malignancies: A national, prospective, and multicentre study. J Infect 2023; 87:46-53. [PMID: 37201859 DOI: 10.1016/j.jinf.2023.05.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 04/28/2023] [Accepted: 05/04/2023] [Indexed: 05/20/2023]
Abstract
OBJECTIVES We describe the current epidemiology, causes, and outcomes of breakthrough invasive fungal infections (BtIFI) in patients with haematologic malignancies. METHODS BtIFI in patients with ≥ 7 days of prior antifungals were prospectively diagnosed (36 months across 13 Spanish hospitals) according to revised EORTC/MSG definitions. RESULTS 121 episodes of BtIFI were documented, of which 41 (33.9%) were proven; 53 (43.8%), probable; and 27 (22.3%), possible. The most frequent prior antifungals included posaconazole (32.2%), echinocandins (28.9%) and fluconazole (24.8%)-mainly for primary prophylaxis (81%). The most common haematologic malignancy was acute leukaemia (64.5%), and 59 (48.8%) patients had undergone a hematopoietic stem-cell transplantation. Invasive aspergillosis, principally caused by non-fumigatus Aspergillus, was the most frequent BtIFI with 55 (45.5%) episodes recorded, followed by candidemia (23, 19%), mucormycosis (7, 5.8%), other moulds (6, 5%) and other yeasts (5, 4.1%). Azole resistance/non-susceptibility was commonly found. Prior antifungal therapy widely determined BtIFI epidemiology. The most common cause of BtIFI in proven and probable cases was the lack of activity of the prior antifungal (63, 67.0%). At diagnosis, antifungal therapy was mostly changed (90.9%), mainly to liposomal amphotericin-B (48.8%). Overall, 100-day mortality was 47.1%; BtIFI was either the cause or an essential contributing factor to death in 61.4% of cases. CONCLUSIONS BtIFI are mainly caused by non-fumigatus Aspergillus, non-albicans Candida, Mucorales and other rare species of mould and yeast. Prior antifungals determine the epidemiology of BtIFI. The exceedingly high mortality due to BtIFI warrants an aggressive diagnostic approach and early initiation of broad-spectrum antifungals different than those previously used.
Collapse
Affiliation(s)
| | | | - Manuela Aguilar-Guisado
- Hospital Universitario Virgen del Rocío, IBIS (Instituto de Biomedicina de Sevilla), Universidad de Sevilla, Sevilla, Spain; Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), (CB21/13/00009), Instituto de Salud Carlos III, Madrid, Spain
| | | | - Júlia Laporte-Amargós
- Hospital Universitari de Bellvitge, IDIBELL (Institut D'Investigació Biomèdica de Bellvitge), Universitat de Barcelona, Barcelona, Spain; Institut Català d'Oncologia, Barcelona, Spain
| | - Marina Machado
- Hospital General Universitario Gregorio Marañón e Instituto de Investigación Sanitaria Gregorio Marañón, Universidad Complutense, Madrid, Spain
| | | | | | | | - Jon Badiola
- Hospital Universitario Virgen de las Nieves, Granada, Spain
| | - Lucia Gómez
- Hospital Universitari Mútua Terrassa, Terrassa, Spain
| | - Isabel Ruiz-Camps
- Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Lucrecia Yáñez
- Hospital Universitario Marqués de Valdecilla-IDIVAL, Santander, Spain
| | | | - Mariana Chumbita
- Hospital Clínic-IDIBAPS, Universitat de Barcelona, Barcelona, Spain
| | - Francesc Marco
- Hospital Clínic-IDIBAPS, Universitat de Barcelona, Barcelona, Spain
| | - Alex Soriano
- Hospital Clínic-IDIBAPS, Universitat de Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), (CB21/13/00009), Instituto de Salud Carlos III, Madrid, Spain
| | - Pedro González
- Hospital Universitario Virgen de las Nieves, Granada, Spain
| | | | | | - Jesús Fortún
- Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Jesús Guinea
- Hospital General Universitario Gregorio Marañón e Instituto de Investigación Sanitaria Gregorio Marañón, Universidad Complutense, Madrid, Spain
| | - Carlota Gudiol
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), (CB21/13/00009), Instituto de Salud Carlos III, Madrid, Spain; Hospital Universitari de Bellvitge, IDIBELL (Institut D'Investigació Biomèdica de Bellvitge), Universitat de Barcelona, Barcelona, Spain; Institut Català d'Oncologia, Barcelona, Spain
| | | | - Maite Ruiz Pérez de Pipaón
- Hospital Universitario Virgen del Rocío, IBIS (Instituto de Biomedicina de Sevilla), Universidad de Sevilla, Sevilla, Spain; Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), (CB21/13/00009), Instituto de Salud Carlos III, Madrid, Spain
| | - Ana Alastruey-Izquierdo
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), (CB21/13/00009), Instituto de Salud Carlos III, Madrid, Spain; Mycology Reference Laboratory, National Centre for Microbiology, Instituto de Salud Carlos III, Majadahonda, Spain
| | - Carolina Garcia-Vidal
- Hospital Clínic-IDIBAPS, Universitat de Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), (CB21/13/00009), Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
8
|
Lynch JP, Zhanel GG. Part 2: Mucormycosis: Focus on Therapy. Expert Rev Anti Infect Ther 2023. [PMID: 37300820 DOI: 10.1080/14787210.2023.2224564] [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: 03/09/2023] [Accepted: 05/25/2023] [Indexed: 06/12/2023]
Abstract
INTRODUCTION Mucormycosis (MCR) a rare but life-threatening infection occurs primarily in immunocompromised hosts. Mortality rates with invasive MCR are high (>30-50%), up to 90% with disseminated disease, but lower (10-30%) with localized cutaneous disease. Due to the rarity of MCR, randomized, controlled therapeutic trials are lacking. Lipid formulations of amphotericin B (LFAB) are the mainstay of therapy but oral triazoles (posaconazole and isavuconazole) may be effective as step-down therapy or in MCR cases refractory to or intolerant of LFAB. Early surgical debridement or excision play important adjunctive roles in localized invasive disease. Control of hyperglycemia in diabetic patients, correction of neutropenia and reduction of immunosuppressive therapy is critical for optimal survival. AREAS COVERED The authors discuss various therapeutic options for mucormycosis. A literature search of mucormycosis therapies was performed via PubMed (up to December 2022), using the key words: invasive fungal infections; mold; mucormycosis; Mucorales; amphotericin B; isavuconazole; posaconazole. EXPERT OPINION Randomized, controlled therapeutic trials are lacking. Lipid formulations of amphotericin B (LFAB) are the mainstay of therapy but oral triazoles (posaconazole and isavuconazole) may be effective as step-down therapy, in MCR cases refractory to or intolerant of LFAB. We encourage early surgical debridement or excision as adjunctive measures.
Collapse
Affiliation(s)
- Joseph P Lynch
- Division of Pulmonary, Critical Care Medicine, Allergy, and Clinical Immunology, Department of Internal Medicine, the David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - George G Zhanel
- Department of Medical Microbiology/Infectious Diseases, Max Rady College of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| |
Collapse
|
9
|
Vallejo C, Jarque I, Fortun J, Casado A, Peman J. IFISTRATEGY: Spanish National Survey of Invasive Fungal Infection in Hemato-Oncologic Patients. J Fungi (Basel) 2023; 9:628. [PMID: 37367564 DOI: 10.3390/jof9060628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 05/18/2023] [Accepted: 05/22/2023] [Indexed: 06/28/2023] Open
Abstract
Recent advances in the treatment of hematologic malignancies have improved the overall survival rate, but the number of patients at risk of developing an invasive fungal infection (IFI) has increased. Invasive infections caused by non-Candida albicans species, non-Aspergillus molds, and azole-resistant Aspergillus fumigatus have been increasingly reported in recent years. We developed a cross-sectional multicenter survey which involved a total of 55 hematologists and infectious disease specialists from a total of 31 Spanish hospitals, to determine the most frequent strategies used for the management of IFIs. Data collection was undertaken through an online survey which took place in 2022. Regarding key strategies, experts usually prefer early treatment for persistent febrile neutropenia, switching to another broad-spectrum antifungal family if azole-resistant Aspergillus is suspected, broad-spectrum azoles and echinocandins as prophylactic treatment in patients receiving midostaurin or venetoclax, and liposomal amphotericin B for breakthrough IFIs after prophylaxis with echinocandins in patients receiving new targeted therapies. For antifungals failing to reach adequate levels during the first days and suspected invasive aspergillosis, the most appropriate strategy would be to associate an antifungal from another family.
Collapse
Affiliation(s)
- Carlos Vallejo
- Hematology Department, Clinic University Hospital of Santiago de Compostela (CHUS), 15706 Santiago de Compostela, Spain
- Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
| | - Isidro Jarque
- Hematology Department, Hospital La Fe, 46026 Valencia, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto Carlos III, 28029 Madrid, Spain
| | - Jesus Fortun
- Infectious Diseases Department, Instituto Ramón y Cajal de Investigación Sanitaria IRYCIS, 28034 Madrid, Spain
- Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), 28805 Madrid, Spain
| | - Araceli Casado
- Pharmacoeconomics and Outcomes Research Iberia (PORIB), 28224 Madrid, Spain
| | - Javier Peman
- Microbiology Department, Hospital La Fe de Valencia, 46026 Valencia, Spain
- Grupo de Investigación Infección Grave, Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain
| |
Collapse
|
10
|
Chen L, Peng W, Lan G, Long G, Yang H, Xu Y, Fu A, Yi H, Wan Q. The application of metagenomic next-generation sequencing in patients with infection or colonization caused by Lichtheimia species. Front Cell Infect Microbiol 2023; 13:1103626. [PMID: 37056706 PMCID: PMC10087083 DOI: 10.3389/fcimb.2023.1103626] [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: 11/20/2022] [Accepted: 03/06/2023] [Indexed: 03/30/2023] Open
Abstract
BackgroundMucormycosis is considered the fourth most common invasive fungal disease after candidiasis, aspergillosis and cryptococcosis. Lichtheimia species accounted for 5%-29% of all mucormycosis. However, available data on species-specific analysis of Lichtheimia infections are limited.MethodsThis study included nine patients hospitalized in five hospitals in two cities in south China with mucormycosis or colonization caused by Lichtheimia species, diagnosed mainly by metagenomic next-generation sequencing (mNGS). The corresponding medical records were reviewed, and the clinical data analyzed included demographic characteristics, site of infection, host factors and type of underlying disease, diagnosis, clinical course, management, and prognosis.ResultsIn this study, nine patients with Lichtheimia infections or colonization had a recent history of haematological malignancy (33.3%), solid organ transplants (33.3%), pulmonary disease (22.2%), and trauma (11.1%) and were categorized as 11.1% (one case) proven, 66.7% (six cases) probable mucormycosis and 22.2% (two cases) colonization. Pulmonary mucormycosis or colonization was the predominant presentation in 77.8% of cases and mucormycosis caused by Lichtheimia resulted in death in four out of seven patients (57.1%).ConclusionThese cases highlight the importance of early diagnosis and combined therapy for these sporadic yet life-threatening infections. Further studies on the diagnosis and control of Lichtheimia infection in China are required.
Collapse
Affiliation(s)
- Lihua Chen
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Weiting Peng
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Gongbin Lan
- Department of Transplant Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Guo Long
- Department of Respiratory and Critical Care Medicine, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Honghui Yang
- Department of Respiratory and Critical Care Medicine, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Yajing Xu
- Department of Hematology, Xiangya Hospital, Central South University, Changsha, China
| | - Ai Fu
- Department of Tuberculosis, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
| | - Huimin Yi
- Department of Surgical Intensive Care Unit, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Qiquan Wan
- Department of Transplant Surgery, The Third Xiangya Hospital, Central South University, Changsha, China
- Engineering and Technology Research Center for Transplantation Medicine of National Health Comission, the Third Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Qiquan Wan,
| |
Collapse
|
11
|
The Mote in Thy Brother's Eyes—Fusarium Solani in Leukemia Host. INFECTIOUS DISEASES IN CLINICAL PRACTICE 2023. [DOI: 10.1097/ipc.0000000000001228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
|
12
|
What Is New in Pulmonary Mucormycosis? J Fungi (Basel) 2023; 9:jof9030307. [PMID: 36983475 PMCID: PMC10057210 DOI: 10.3390/jof9030307] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 02/14/2023] [Accepted: 02/16/2023] [Indexed: 03/06/2023] Open
Abstract
Mucormycosis is a rare but life-threatening fungal infection due to molds of the order Mucorales. The incidence has been increasing over recent decades. Worldwide, pulmonary mucormycosis (PM) presents in the lungs, which are the third main location for the infection after the rhino-orbito-cerebral (ROC) areas and the skin. The main risk factors for PM include hematological malignancies and solid organ transplantation, whereas ROC infections are classically favored by diabetes mellitus. The differences between the ROC and pulmonary locations are possibly explained by the activation of different mammalian receptors—GRP78 in nasal epithelial cells and integrin β1 in alveolar epithelial cells—in response to Mucorales. Alveolar macrophages and neutrophils play a key role in the host defense against Mucorales. The diagnosis of PM relies on CT scans, cultures, PCR tests, and histology. The reversed halo sign is an early, but very suggestive, sign of PM in neutropenic patients. Recently, the serum PCR test showed a very encouraging performance for the diagnosis and follow-up of mucormycosis. Liposomal amphotericin B is the drug of choice for first-line therapy, together with correction of underlying disease and surgery when feasible. After a stable or partial response, the step-down treatment includes oral isavuconazole or posaconazole delayed release tablets until a complete response is achieved. Secondary prophylaxis should be discussed when there is any risk of relapse, such as the persistence of neutropenia or the prolonged use of high-dose immunosuppressive therapy. Despite these novelties, the mortality rate from PM remains higher than 50%. Therefore, future research must define the place for combination therapy and adjunctive treatments, while the development of new treatments is necessary.
Collapse
|
13
|
Aerts R, Bevers S, Beuselinck K, Schauwvlieghe A, Lagrou K, Maertens J. Blood Mucorales PCR to track down Aspergillus and Mucorales co-infections in at-risk hematology patients: A case-control study. Front Cell Infect Microbiol 2022; 12:1080921. [PMID: 36569194 PMCID: PMC9774025 DOI: 10.3389/fcimb.2022.1080921] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 11/21/2022] [Indexed: 12/13/2022] Open
Abstract
Introduction Serum Mucorales PCR can precede the final diagnosis of invasive mucormycosis by several days or weeks and could therefore be useful as a non-invasive screening tool. Methods We assessed the performance of a commercial Mucorales PCR assay (MucorGenius®, PathoNostics, Maastricht, The Netherlands) on prospectively collected banked sera from hematology patients at risk for invasive mould infections. We evaluated if there is an underestimated incidence of missed Mucorales co-infections in patients with invasive aspergillosis (IA). We tested Mucorales PCR on the sera of all patients with a diagnosis of at least possible IA (EORTC-MSGERC consensus criteria) before the start of any antifungal therapy, and in a control group of similar high-risk hematology patients without IA (in a 1:4 ratio). When a positive Mucorales PCR was observed, at least 5 serum samples taken before and after the positive one were selected. Results Mucorales PCR was performed in 46 diagnostic serum samples of cases and in 184 controls. Serum Mucorales PCR was positive in 4 cases of IA (8.7%; 12.9% of probable cases) and in 1 control case (0.5%) (p=0.0061, OR=17.43 (1.90-159.96). Post-mortem cultures of the positive control became positive for Rhizopus arrhizus. Mortality of IA cases with and without a positive Mucorales PCR was not significantly different. Only in the PCR positive control case, serial serum samples before and after the diagnostic sample were also positive. Discussion It is not entirely clear what a positive Mucorales PCR in these cases implies since the 4 Mucorales PCR positive cases were treated with antifungals with activity against Mucorales. In addition, PCR was positive only once. This study does not provide enough evidence to implement Mucorales PCR screening. However, our findings emphasize once more the importance of considering the possibility of dual mould infections, even in patients with a positive galactomannan detection.
Collapse
Affiliation(s)
- Robina Aerts
- Department of Internal Medicine, University Hospitals Leuven, Leuven, Belgium,Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium,*Correspondence: Robina Aerts,
| | - Sien Bevers
- Department of Haematology, University Hospitals Leuven, Leuven, Belgium
| | - Kurt Beuselinck
- Department of Laboratory Medicine, University Hospitals Leuven, Leuven, Belgium
| | | | - Katrien Lagrou
- Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium,Department of Laboratory Medicine, University Hospitals Leuven, Leuven, Belgium
| | - Johan Maertens
- Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium,Department of Haematology, University Hospitals Leuven, Leuven, Belgium
| |
Collapse
|
14
|
High incidence of resistant breakthrough invasive fungal infections (IFD) in patients treated for acute gastrointestinal graft-versus-host disease (GI GVHD) following allogeneic haematopoietic cell transplantation. Bone Marrow Transplant 2022; 57:1712-1715. [PMID: 35970876 DOI: 10.1038/s41409-022-01773-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 07/26/2022] [Accepted: 07/28/2022] [Indexed: 11/08/2022]
|
15
|
Taynton T, Barlow G, Allsup D. PRO: Biomarker surveillance for invasive fungal infections without antifungal prophylaxis could safely reduce antifungal use in acute leukaemia. JAC Antimicrob Resist 2022; 4:dlac074. [PMID: 35873180 PMCID: PMC9305519 DOI: 10.1093/jacamr/dlac074] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 06/07/2022] [Indexed: 11/14/2022] Open
Abstract
Mould-active antifungal prophylaxis is frequently used to prevent invasive fungal infection in patients with acute leukaemia being treated with intensive chemotherapy. Invasive fungal infections are difficult to diagnose, and despite the use of prophylaxis a high proportion of patients still receive therapeutic antifungals. Antifungal medications have important interactions, can cause serious adverse events, and may drive the proliferation of antifungal resistance. The use of two biomarkers, such as galactomannan in combination with the less-specific β-d-glucan, can mitigate the risk of not detecting non-Aspergillus species, as well as improving pooled sensitivity and specificity. We argue that regular biomarkers could be used safely as part of an antifungal stewardship strategy to reduce antifungal use, by both screening for infection in patients not on prophylaxis and ruling out infection in patients treated empirically.
Collapse
Affiliation(s)
- Thomas Taynton
- Hull University Teaching Hospitals NHS Trust, Castle Hill
Hospital, Castle Road, Cottingham, Hull, HU16
5JQ, UK
- Centre for Atherothrombosis and Metabolic Disease, Hull York Medical
School, University of Hull, Hull, HU6 7RX,
UK
| | - Gavin Barlow
- Hull University Teaching Hospitals NHS Trust, Castle Hill
Hospital, Castle Road, Cottingham, Hull, HU16
5JQ, UK
- Hull York Medical School, University of York, Heslington,
York, YO10 5DD, UK
| | - David Allsup
- Hull University Teaching Hospitals NHS Trust, Castle Hill
Hospital, Castle Road, Cottingham, Hull, HU16
5JQ, UK
- Centre for Atherothrombosis and Metabolic Disease, Hull York Medical
School, University of Hull, Hull, HU6 7RX,
UK
| |
Collapse
|
16
|
Machherndl-Spandl S, Vockenhuber T, Binder M, Weltermann A, Apfalter P, Lass-Flörl C, Girschikofsky M. Efficacy and safety of voriconazole as invasive fungal infection prophylaxis in patients with acute myeloid leukemia. Leuk Lymphoma 2022; 63:2330-2335. [DOI: 10.1080/10428194.2022.2068006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- Sigrid Machherndl-Spandl
- Department of Internal Medicine I (Hematology with Stem Cell Transplantation, Hemostaseology and Medical Oncology), Ordensklinikum Linz Elisabethinen, Linz, Austria
| | - Thomas Vockenhuber
- Department of Internal Medicine I (Hematology with Stem Cell Transplantation, Hemostaseology and Medical Oncology), Ordensklinikum Linz Elisabethinen, Linz, Austria
| | - Michaela Binder
- Department of Internal Medicine I (Hematology with Stem Cell Transplantation, Hemostaseology and Medical Oncology), Ordensklinikum Linz Elisabethinen, Linz, Austria
| | - Ansgar Weltermann
- Department of Internal Medicine I (Hematology with Stem Cell Transplantation, Hemostaseology and Medical Oncology), Ordensklinikum Linz Elisabethinen, Linz, Austria
| | - Petra Apfalter
- Institute of Hygiene, Microbiology and Tropical Medicine and analyse BioLab, National Reference Centre for Antimicrobial Resistance, IZIM – Ordensklinikum Linz Elisabethinen, Linz, Austria
| | - Cornelia Lass-Flörl
- Division of Hygiene and Medical Microbiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Michael Girschikofsky
- Department of Internal Medicine I (Hematology with Stem Cell Transplantation, Hemostaseology and Medical Oncology), Ordensklinikum Linz Elisabethinen, Linz, Austria
| |
Collapse
|
17
|
Weber JK, Scharf S, Walther G, Flüh G, MacKenzie CR, Kondakci M, Henrich B, Kohns Vasconcelos M. Detection of invasive Trichosporon asahii in patient blood by a fungal PCR array. Access Microbiol 2022; 3:000285. [PMID: 35024550 PMCID: PMC8749139 DOI: 10.1099/acmi.0.000285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 09/30/2021] [Indexed: 11/24/2022] Open
Abstract
Rare invasive fungal infections are increasingly emerging in hosts with predisposing factors such as immunodeficiency. Their timely diagnosis remains difficult, as their clinical picture may initially mimic infections with more common fungal species and species identification may be difficult with routine methods or may require time-consuming subcultures. This often results in ineffective drug administration and fatal outcomes. We report on a patient in their early twenties with mixed cellularity classical Hodgkin lymphoma with a disseminated Trichosporon asahii (T. asahii) infection. Even though pathogen detection and identification was possible via the standard procedure consisting of culture followed by matrix-assisted laser desorption ionisation–time of flight (MALDI-TOF) mass spectrometry, the patient passed away in the course of multi organ failure. Herein, we report on a retrospectively applied experimental diagnostic fungal PCR-analysis used on an EDTA blood sample and consisting of two pan-fungal reactions and seven branch-specific reactions. Regarding invasive T. asahii infection, this PCR array could considerably shorten time to diagnosis and switch to a targeted therapy with triazoles.
Collapse
Affiliation(s)
- Jasmin K Weber
- Institute for Medical Microbiology and Hospital Hygiene, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Sebastian Scharf
- Institute for Medical Microbiology and Hospital Hygiene, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Grit Walther
- German National Reference Centre for Invasive Fungal Infections, Leibniz Institute for Natural Product Research and Infection Biology-Hans Knöll Institute, Jena, Germany
| | - Greta Flüh
- Institute for Medical Microbiology and Hospital Hygiene, Heinrich Heine University Düsseldorf, Düsseldorf, Germany.,Labor Dr. Wisplinghoff, Cologne, Germany
| | - Colin R MacKenzie
- Institute for Medical Microbiology and Hospital Hygiene, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Mustafa Kondakci
- Department of Haematology, Oncology and Clinical Immunology, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Birgit Henrich
- Institute for Medical Microbiology and Hospital Hygiene, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Malte Kohns Vasconcelos
- Institute for Medical Microbiology and Hospital Hygiene, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
18
|
Sannathimmappa MB, Nambiar V, Aravindakshan R. Storm of a rare opportunistic life threatening mucormycosis among post COVID-19 patients: A tale of two pathogens. Int J Crit Illn Inj Sci 2022; 12:38-46. [PMID: 35433396 PMCID: PMC9008285 DOI: 10.4103/ijciis.ijciis_48_21] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 07/12/2021] [Accepted: 09/01/2021] [Indexed: 11/22/2022] Open
Abstract
Mucormycosis is a rare but life-threatening opportunistic fungal infection caused by a group of molds that belong to Zygomycetes of the order Mucorales. These fungi are found in the environment such as soil, decaying vegetation, and organic matters. Sporangiospores present in the environment enter the human body through inhalation or direct skin inoculation by trauma or ingestion and result in pulmonary, cutaneous, and gastrointestinal mucormycosis, respectively, in immunocompromised hosts. Patients with uncontrolled diabetes, hematological malignancies, high-dose glucocorticoid therapy, iron overload, and organ transplantation are at high risk of acquiring mucormycosis. The second wave of severe acute respiratory syndrome coronavirus 2 [SARS-CoV-2] affected India severely with the highest number of cases and deaths compared to all other countries. Additionally, the country was affected by emergence of rare but life-threatening mucormycosis. Currently, many coronavirus disease 2019 patients with underlying risk factors such as uncontrolled diabetes, high-dose steroid therapy, and exposure to mechanical ventilation have developed mucormycosis. Inhalation is the most common mode of transmission that results in colonization of sporangiospores in the nose. In immunocompromised host, sporangiospores germinate, and subsequently form hyphae. These hyphae invade into tissues, and produce tissue infarction, necrosis, and thrombosis. Angioinvasion causes hematogenous dissemination to many organs, predominantly to brain, that result in rhino-orbital-cerebral mucormycosis. Clinical characteristics, radio imaging, fungal culture, histopathology, and molecular techniques are the key diagnostic methods. Surgical intervention and aggressive antifungal therapy are the main management strategies. Amphotericin B is the drug of choice for treatment of mucormycosis, whereas posaconazole or isavuconazole is used for step-down therapy and salvage therapy.
Collapse
Affiliation(s)
- Mohan Bilikallahalli Sannathimmappa
- Department of Microbiology, College of Medicine and Health Sciences, National University of Science and Technology, Sohar Campus, Andhra Pradesh, India
- Department of Microbiology, National University of Science and Technology, Sohar Campus, Sultanate of Oman
| | - Vinod Nambiar
- Department of Microbiology, College of Medicine and Health Sciences, National University of Science and Technology, Sohar Campus, Andhra Pradesh, India
- Department of Microbiology, National University of Science and Technology, Sohar Campus, Sultanate of Oman
| | - Rajeev Aravindakshan
- Department of Community Medicine, All India Institute of Medical Sciences, Mangalagiri, Andhra Pradesh, India
| |
Collapse
|
19
|
Khanina A, Tio SY, Ananda‐Rajah MR, Kidd SE, Williams E, Chee L, Urbancic K, Thursky KA. Consensus guidelines for antifungal stewardship, surveillance and infection prevention, 2021. Intern Med J 2021; 51 Suppl 7:18-36. [DOI: 10.1111/imj.15586] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Anna Khanina
- National Centre for Infections in Cancer Peter MacCallum Cancer Centre Melbourne Victoria Australia
- Sir Peter MacCallum Department of Oncology The University of Melbourne Melbourne Victoria Australia
| | - Shio Yen Tio
- National Centre for Infections in Cancer Peter MacCallum Cancer Centre Melbourne Victoria Australia
- Sir Peter MacCallum Department of Oncology The University of Melbourne Melbourne Victoria Australia
| | - Michelle R. Ananda‐Rajah
- Department of General Medicine Alfred Health Melbourne Victoria Australia
- Department of Infectious Diseases Alfred Health Melbourne Victoria Australia
| | - Sarah E. Kidd
- National Mycology Reference Centre Microbiology and Infectious Diseases, SA Pathology Adelaide South Australia Australia
- School of Biological Sciences University of Adelaide Adelaide South Australia Australia
| | - Eloise Williams
- Department of Microbiology Royal Melbourne Hospital Melbourne Victoria Australia
- Department of Microbiology and Immunology The Peter Doherty Institute for Immunity and Infection, The University of Melbourne Melbourne Parkville Victoria Australia
| | - Lynette Chee
- Department of Clinical Haematology Peter MacCallum Cancer Centre and Royal Melbourne Hospital Melbourne Victoria Australia
- Department of Medicine The University of Melbourne Melbourne Victoria Australia
| | - Karen Urbancic
- National Centre for Infections in Cancer Peter MacCallum Cancer Centre Melbourne Victoria Australia
- Department of Medicine The University of Melbourne Melbourne Victoria Australia
- Pharmacy Department Austin Health Melbourne Victoria Australia
- National Centre for Antimicrobial Stewardship Melbourne Victoria Australia
| | - Karin A. Thursky
- National Centre for Infections in Cancer Peter MacCallum Cancer Centre Melbourne Victoria Australia
- Department of Medicine The University of Melbourne Melbourne Victoria Australia
- National Centre for Antimicrobial Stewardship Melbourne Victoria Australia
- Department of Infectious Diseases Peter MacCallum Cancer Centre Melbourne Victoria Australia
- Victorian Infectious Diseases Service The Peter Doherty Institute for Immunity and Infection, Royal Melbourne Hospital Melbourne Victoria Australia
| | | |
Collapse
|
20
|
Changing Epidemiology of Invasive Fungal Disease in Allogeneic Hematopoietic Stem Cell Transplantation. J Fungi (Basel) 2021; 7:jof7100848. [PMID: 34682269 PMCID: PMC8539090 DOI: 10.3390/jof7100848] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 10/05/2021] [Accepted: 10/07/2021] [Indexed: 12/23/2022] Open
Abstract
Invasive fungal disease (IFD) is a common cause of morbidity and mortality in patients with hematologic malignancies, especially among those undergoing allogeneic hematopoietic stem cell transplantation (HSCT). The epidemiology of IFD in HSCT patients has been evolving over the last decades, mainly in relation to changes in HSCT therapies such as antifungal prophylaxis. A progressive decrease in Candida albicans infection has been documented, alongside a progressive increase in infections caused by non-albicans Candida species, filamentous fungi, and/or multidrug-resistant fungi. Currently, the most frequent IFD is invasive aspergillosis. In some parts of the world, especially in north Central Europe, a high percentage of Aspergillus fumigatus isolates are azole-resistant. New diagnostic techniques have documented the existence of cryptic Aspergillus species with specific characteristics. An increase in mucormycosis and fusariosis diagnoses, as well as diagnoses of other rare fungi, have also been described. IFD epidemiology is likely to continue changing further due to both an increased use of mold-active antifungals and a lengthened survival of patients with HSCT that may result in hosts with weaker immune systems. Improvements in microbiology laboratories and the widespread use of molecular diagnostic tools will facilitate more precise descriptions of current IFD epidemiology. Additionally, rising resistance to antifungal drugs poses a major threat. In this scenario, knowledge of current epidemiology and accurate IFD diagnoses are mandatory in order to establish correct prophylaxis guidelines and appropriate early treatments.
Collapse
|
21
|
Invasive Fungal Disease in Patients with Newly Diagnosed Acute Myeloid Leukemia. J Fungi (Basel) 2021; 7:jof7090761. [PMID: 34575799 PMCID: PMC8471241 DOI: 10.3390/jof7090761] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 09/07/2021] [Accepted: 09/09/2021] [Indexed: 01/18/2023] Open
Abstract
This single-center retrospective study of invasive fungal disease (IFD) enrolled 251 adult patients undergoing induction chemotherapy for newly diagnosed acute myeloid leukemia (AML) from 2014–2019. Patients had primary AML (n = 148, 59%); antecedent myelodysplastic syndrome (n = 76, 30%), or secondary AML (n = 27, 11%). Seventy-five patients (30%) received an allogeneic hematopoietic cell transplant within the first year after induction chemotherapy. Proven/probable IFD occurred in 17 patients (7%). Twelve of the 17 (71%) were mold infections, including aspergillosis (n = 6), fusariosis (n = 3), and mucomycosis (n = 3). Eight breakthrough IFD (B-IFD), seven of which were due to molds, occurred in patients taking antifungal prophylaxis. Patients with proven/probable IFD had a significantly greater number of cumulative neutropenic days than those without an IFD, HR = 1.038 (95% CI 1.018–1.059), p = 0.0001. By cause-specific proportional hazards regression, the risk for IFD increased by 3.8% for each day of neutropenia per 100 days of follow up. Relapsed/refractory AML significantly increased the risk for IFD, HR = 7.562 (2.585–22.123), p = 0.0002, and Kaplan-Meier analysis showed significantly higher mortality at 1 year in patients who developed a proven/probable IFD, p = 0.02. IFD remains an important problem among patients with AML despite the use of antifungal prophylaxis, and development of IFD is associated with increased mortality in these patients.
Collapse
|
22
|
Chen X, Wang J, Wang S, Jin J, Li J, Gao S, Li J, Li J, Liu Q, Hu Y, Lin D, Sun Z, Yang J, Hu J, Wu X, Huang X, Shao Z, Deng Q, Wang C, Liu L, Chen H, Wang J, Wei X, Shen J, Zhang X, Wu D. Real-world assessment of the effectiveness of posaconazole for the prophylaxis and treatment of invasive fungal infections in hematological patients: A retrospective observational study. Medicine (Baltimore) 2021; 100:e26772. [PMID: 34397725 PMCID: PMC8322488 DOI: 10.1097/md.0000000000026772] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 07/04/2021] [Indexed: 01/04/2023] Open
Abstract
The aim of the study was to analyze the efficacy of posaconazole for the prophylaxis and treatment of invasive fungal diseases (IFDs) in patients with hematological malignancies.In this retrospective observational multi-center study, 762 patients from 25 Chinese hematological centers were enrolled. Inclusion criteria were patients with hematological malignancy or they had undergone hematopoietic stem cell transplantation and received at least 1 dose of posaconazole. The primary endpoints were the observation of breakthrough rates and the clinical efficacy of posaconazole prophylaxis. The secondary endpoint was the efficacy of posaconazole for the treatment of IFDs.Of the 762 enrolled patients, 456 (59.8%) were prescribed posaconazole prophylactically while 243 (31.9%) received posaconazole as an IFD treatment (12 proven, 61 probable, 109 possible, and 61 unclassified IFD cases) for ≥7 days. The overall IFD breakthrough rate (probable cases) for the ≥4 days prophylactic treatment (n = 445) group was 1.6% (95% Cl: 0.6%-3.2%), with breakthrough rates of 2.6% for acute myeloid leukemia/myelodysplastic syndrome patients undergoing chemotherapy and 2.2% for hematopoietic stem cell transplantation patients. For primary antifungal prophylaxis, the breakthrough rate was 1.9% and for secondary antifungal prophylaxis 0%. The overall effective IFD remission rate of patients treated for ≥7 days with posaconazole was 56.0% and the effective remission rate of proven/probable/possible IFD cases was 59.3%. The effective remission rate of posaconazole as salvage therapy was 50% (95% CI: 32.4%-67.6%) including 75% (CI: 19.4%-99.4%) for Aspergillus infections.The present retrospective study confirmed posaconazole as IFD prophylaxis and medication for hematological malignancy patients undergoing various treatments in China.
Collapse
Affiliation(s)
- Xiaochen Chen
- Department of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Jianxiang Wang
- Leukemia Diagnosis and Treatment Center, Blood Diseases Hospital, Chinese Academy of Medical Sciences, Tianjin, China
| | - Sanbin Wang
- Department of Hematology, Kunming General Hospital, Chengdu Military Region, Kunming, Yunnan Province, China
| | - Jie Jin
- Department of Hematology, The First Affiliated Hospital of Medical School of Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Junmin Li
- Department of Hematology, Ruijin Hospital Affiliated to Medical College of Shanghai Jiaotong University, Shanghai, China
| | - Sujun Gao
- Department of Hematology, The First Bethune Hospital of Jilin University, Changchun, Jilin Province, China
| | - Jianyong Li
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Juan Li
- Department of Hematology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong Province, China
| | - Qifa Liu
- Department of Hematology, Nanfang Hospital Affiliated to Southern Medical University, Guangzhou, Guangdong Province, China
| | - Yu Hu
- Department of Hematology, Wuhan Union Hospital Affiliated to Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Dongjun Lin
- Department of Hematology, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong Province, China
| | - Zimin Sun
- Department of Hematology, Anhui Provincial Hospital, The First Affiliated Hospital of USTC, Hefei, Anhui Province, China
| | - Jianmin Yang
- Department of Hematology, Changhai Hospital, Shanghai, China
| | - Jianda Hu
- Department of Hematology, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
| | - Xiaoxiong Wu
- Department of Hematology, Fourth Medical Center of PLA General Hospital, Beijing, China
| | - Xiaojun Huang
- Department of Hematology, Peking University People's Hospital, Beijing, China
| | - Zonghong Shao
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, China
| | - Qi Deng
- Department of Hematology, Tianjin First Central Hospital, Tianjin, China
| | - Chun Wang
- Department of Hematology, Shanghai General Hospital, Shanghai, China
| | - Li Liu
- Department of Hematology, Tangdu Hospital, Xi’an, Shaanxi Province, China
| | - Hu Chen
- Department of Hematopoietic Stem Cell Transplantation, 307 Hospital of PLA, Beijing, China
| | - Jingbo Wang
- Department of Hematology, Aerospace Central Hospital, Beijing, China
| | - Xudong Wei
- Department of Hematology, Henan Cancer Hospital, Zhengzhou, Henan Province, China
| | - Jianping Shen
- Department of Hematology, Zhejiang Provincial Hospital of TCM, Hangzhou. Zhejiang Province, China
| | - Xi Zhang
- Department of Hematology, Xinqiao Hospital, Chongqing, China
| | - Depei Wu
- Department of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| |
Collapse
|
23
|
Vehreschild JJ, Koehler P, Lamoth F, Prattes J, Rieger C, Rijnders BJA, Teschner D. Future challenges and chances in the diagnosis and management of invasive mould infections in cancer patients. Med Mycol 2021; 59:93-101. [PMID: 32898264 PMCID: PMC7779224 DOI: 10.1093/mmy/myaa079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 07/31/2020] [Accepted: 08/18/2020] [Indexed: 11/15/2022] Open
Abstract
Diagnosis, treatment, and management of invasive mould infections (IMI) are challenged by several risk factors, including local epidemiological characteristics, the emergence of fungal resistance and the innate resistance of emerging pathogens, the use of new immunosuppressants, as well as off-target effects of new oncological drugs. The presence of specific host genetic variants and the patient's immune system status may also influence the establishment of an IMI and the outcome of its therapy. Immunological components can thus be expected to play a pivotal role not only in the risk assessment and diagnosis, but also in the treatment of IMI. Cytokines could improve the reliability of an invasive aspergillosis diagnosis by serving as biomarkers as do serological and molecular assays, since they can be easily measured, and the turnaround time is short. The use of immunological markers in the assessment of treatment response could be helpful to reduce overtreatment in high risk patients and allow prompt escalation of antifungal treatment. Mould-active prophylaxis could be better targeted to individual host needs, leading to a targeted prophylaxis in patients with known immunological profiles associated with high susceptibility for IMI, in particular invasive aspergillosis. The alteration of cellular antifungal immune response through oncological drugs and immunosuppressants heavily influences the outcome and may be even more important than the choice of the antifungal treatment. There is a need for the development of new antifungal strategies, including individualized approaches for prevention and treatment of IMI that consider genetic traits of the patients. Lay Abstract Anticancer and immunosuppressive drugs may alter the ability of the immune system to fight invasive mould infections and may be more important than the choice of the antifungal treatment. Individualized approaches for prevention and treatment of invasive mold infections are needed.
Collapse
Affiliation(s)
- Jörg Janne Vehreschild
- Department of Internal Medicine, Hematology, and Oncology, University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt am Main, Germany; Department I for Internal Medicine, University Hospital of Cologne, Cologne, Germany; German Centre for Infection Research, partner site Bonn-Cologne, University of Cologne, Cologne, Germany
| | - Philipp Koehler
- 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), Excellence Center for Medical Mycology (ECMM), Cologne, Germany.,University of Cologne, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne, Germany
| | - Frédéric Lamoth
- Infectious Diseases Service, Department of Medicine, Lausanne University Hospital, Lausanne, Switzerland.,Institute of Microbiology, Department of Laboratories, Lausanne University Hospital, Lausanne, Switzerland
| | - Juergen Prattes
- Section of Infectious Diseases and Tropical Medicine, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | | | - Bart J A Rijnders
- Internal Medicine and Infectious Diseases, Erasmus MC University Medical Center, Rotterdam, Netherlands
| | - Daniel Teschner
- Department of Hematology, Medical Oncology, and Pneumology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| |
Collapse
|
24
|
Abstract
Introduction: Invasive fungal infection carries a high morbidity, mortality and economic cost. In recent times, a rising incidence of fungal infection and antifungal resistance is occurring which has prompted the development of novel antifungal agents.Areas covered:In this perspective, the authors describe the current status of registered antifungals and their limitations in the treatment of invasive fungal infection. They also go on to describe the new antifungal agents that are in the clinical stage of development and how they might be best utilized in patient care in the future.Expert opinion: The antifungal drug development pipeline has responded to a growing need for new agents to effectively treat fungal disease without concomitant toxicity or issues with drug tolerance. Olorofim (F901318), ibrexafungerp (SCY-078), fosmanogepix (APX001), rezafungin (CD101), oteseconazole (VT-1161), encochleated amphotericin B (MAT2203), nikkomycin Z (NikZ) and ATI-2307 are all in the clinical stage of development and offer great promise in offering clinicians better agents to treat these difficult infections.
Collapse
Affiliation(s)
- Adam G Stewart
- Department of Infectious Diseases, Royal Brisbane and Women's Hospital, Brisbane, Australia.,Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Royal Brisbane and Women's Hospital Campus, Brisbane, Australia
| | - David L Paterson
- Department of Infectious Diseases, Royal Brisbane and Women's Hospital, Brisbane, Australia.,Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Royal Brisbane and Women's Hospital Campus, Brisbane, Australia
| |
Collapse
|
25
|
Martín Gómez MT, Salavert Lletí M. [Mucormycosis: Current and future management perspective]. Rev Iberoam Micol 2021; 38:91-100. [PMID: 34144835 DOI: 10.1016/j.riam.2021.04.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 03/20/2021] [Accepted: 04/08/2021] [Indexed: 02/06/2023] Open
Abstract
Infections caused by mucorales, with an increasing incidence after candidiasis and aspergillosis, are characterized by the fast angioinvasion of blood vessels and invasion of neighboring organs or structures. Mucorales most commonly cause rhinocerebral, pulmonary, cutaneous, digestive or disseminated infections, and their spread is favored by certain underlying diseases (diabetes, kidney failure) and risk factors (neutropenia, immunosuppression, iron overload). These infections have a high mortality rate, over 40% in many series, and the key to their cure depends on both an early diagnosis and an antifungal treatment, associated in most cases with extensive surgical debridement and other adjunctive therapies. Currently, there are international guidelines, not only local ones, for the management of mucormycosis, in which it is considered by consensus and with a strong recommendation that first-line treatment with high-dose liposomal amphotericin B is the best choice. The combined antifungal treatment of polyene agents with triazoles or candins remains in open debate.
Collapse
Affiliation(s)
- María Teresa Martín Gómez
- Sección de Micología, Servicio de Microbiología, Hospital Universitario Vall d'Hebron, Barcelona, España
| | - Miguel Salavert Lletí
- Unidad de Enfermedades Infecciosas (Área Clínica Médica), Hospital Universitario y Politécnico La Fe, Valencia, España.
| |
Collapse
|
26
|
Rapidly expanded partially HLA DRB1-matched fungus-specific T cells mediate in vitro and in vivo antifungal activity. Blood Adv 2021; 4:3443-3456. [PMID: 32722785 DOI: 10.1182/bloodadvances.2020001565] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 06/24/2020] [Indexed: 01/23/2023] Open
Abstract
Invasive fungal infections are a major cause of disease and death in immunocompromised hosts, including patients undergoing allogeneic hematopoietic stem cell transplant (HSCT). Recovery of adaptive immunity after HSCT correlates strongly with recovery from fungal infection. Using initial selection of lymphocytes expressing the activation marker CD137 after fungal stimulation, we rapidly expanded a population of mainly CD4+ T cells with potent antifungal characteristics, including production of tumor necrosis factor α, interferon γ, interleukin-17, and granulocyte-macrophage colony stimulating factor. Cells were manufactured using a fully good manufacturing practice-compliant process. In vitro, the T cells responded to fungal antigens presented on fully and partially HLA-DRB1 antigen-matched presenting cells, including when the single common DRB1 antigen was allelically mismatched. Administration of antifungal T cells lead to reduction in the severity of pulmonary and cerebral infection in an experimental mouse model of Aspergillus. These data support the establishment of a bank of cryopreserved fungus-specific T cells using normal donors with common HLA DRB1 molecules and testing of partially HLA-matched third-party donor fungus-specific T cells as a potential therapeutic in patients with invasive fungal infection after HSCT.
Collapse
|
27
|
García-Vidal C, Vázquez L, Jarque I. [Relevance of liposomal amphotericin B in the treatment of invasive fungal infections in patients with hematologic malignancies]. Rev Iberoam Micol 2021; 38:61-67. [PMID: 33994104 DOI: 10.1016/j.riam.2021.03.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 03/18/2021] [Accepted: 03/25/2021] [Indexed: 12/20/2022] Open
Abstract
Liposomal amphotericin B (L-AmB) has been a key cornerstone for the management of invasive fungal infections (IFI) caused by a wide array of molds and yeasts during the last three decades. Multiple studies performed over this period have generated a large body of evidence on its efficacy and safety, becoming the main antifungal agent in the management of IFI in patients with hematologic malignancies in several not mutually exclusive clinical settings. First, L-AmB is the most commonly used antifungal agent in patients undergoing intensive chemotherapy for acute leukemia and high-risk myelodysplastic syndrome, as well as in hematopoietic stem cell transplant recipients. Additionally, due to the administration of newer targeted therapies (such as monoclonal antibodies or small molecule inhibitors), opportunistic mold infections are increasingly being reported in patients with hematologic malignancies usually considered low-risk for IFI. These agents usually have a high drug-drug interaction potential, being triazoles, commonly used for antifungal prophylaxis, included. Finally, patients developing breakthrough IFI because of either subtherapeutic concentrations of antifungal prophylactic drugs in blood or selection of resistant strains, require broad spectrum antifungal therapy, usually with an antifungal of a different class. In both situations, L-AmB remains as the best option for early antifungal therapy.
Collapse
Affiliation(s)
| | - Lourdes Vázquez
- Servicio de Hematología, Hospital Universitario, Salamanca, España
| | - Isidro Jarque
- Servicio de Hematología, Hospital Universitario y Politécnico La Fe, Valencia, España.
| |
Collapse
|
28
|
Alegria W, Patel PK. The Current State of Antifungal Stewardship in Immunocompromised Populations. J Fungi (Basel) 2021; 7:352. [PMID: 33946217 PMCID: PMC8145600 DOI: 10.3390/jof7050352] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 04/24/2021] [Accepted: 04/26/2021] [Indexed: 01/05/2023] Open
Abstract
Inappropriate antifungal use is prevalent and can lead to drug-resistant fungi, expose patients to adverse drug events, and increase healthcare costs. While antimicrobial stewardship programs have traditionally focused on antibiotic use, the need for targeted antifungal stewardship (AFS) intervention has garnered interest in recent years. Despite this, data on AFS in immunocompromised patient populations is limited. This paper will review the current state of AFS in this complex population and explore opportunities for multidisciplinary collaboration.
Collapse
Affiliation(s)
- William Alegria
- Department of Quality, Patient Safety and Effectiveness, Stanford Health Care, 300 Pasteur Drive, Lane 134 L1C36, Stanford, CA 94305, USA
- Stanford Antimicrobial Safety and Sustainability Program, Stanford, CA 94305, USA
| | - Payal K. Patel
- Division of Infectious Diseases, Department of Internal Medicine, Ann Arbor VA Medical Center, Ann Arbor, MI 48105, USA;
- Division of Infectious Diseases, Department of Internal Medicine, University of Michigan, 2215 Fuller Rd, Ann Arbor, MI 48105, USA
| |
Collapse
|
29
|
Stanford FA, Matthies N, Cseresnyés Z, Figge MT, Hassan MIA, Voigt K. Expression Patterns in Reductive Iron Assimilation and Functional Consequences during Phagocytosis of Lichtheimia corymbifera, an Emerging Cause of Mucormycosis. J Fungi (Basel) 2021; 7:jof7040272. [PMID: 33916756 PMCID: PMC8065604 DOI: 10.3390/jof7040272] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 03/17/2021] [Accepted: 03/29/2021] [Indexed: 12/30/2022] Open
Abstract
Iron is an essential micronutrient for most organisms and fungi are no exception. Iron uptake by fungi is facilitated by receptor-mediated internalization of siderophores, heme and reductive iron assimilation (RIA). The RIA employs three protein groups: (i) the ferric reductases (Fre5 proteins), (ii) the multicopper ferroxidases (Fet3) and (iii) the high-affinity iron permeases (Ftr1). Phenotyping under different iron concentrations revealed detrimental effects on spore swelling and hyphal formation under iron depletion, but yeast-like morphology under iron excess. Since access to iron is limited during pathogenesis, pathogens are placed under stress due to nutrient limitations. To combat this, gene duplication and differential gene expression of key iron uptake genes are utilized to acquire iron against the deleterious effects of iron depletion. In the genome of the human pathogenic fungus L. corymbifera, three, four and three copies were identified for FRE5, FTR1 and FET3 genes, respectively. As in other fungi, FET3 and FTR1 are syntenic and co-expressed in L. corymbifera. Expression of FRE5, FTR1 and FET3 genes is highly up-regulated during iron limitation (Fe-), but lower during iron excess (Fe+). Fe- dependent upregulation of gene expression takes place in LcFRE5 II and III, LcFTR1 I and II, as well as LcFET3 I and II suggesting a functional role in pathogenesis. The syntenic LcFTR1 I–LcFET3 I gene pair is co-expressed during germination, whereas LcFTR1 II- LcFET3 II is co-expressed during hyphal proliferation. LcFTR1 I, II and IV were overexpressed in Saccharomyces cerevisiae to represent high and moderate expression of intracellular transport of Fe3+, respectively. Challenge of macrophages with the yeast mutants revealed no obvious role for LcFTR1 I, but possible functions of LcFTR1 II and IVs in recognition by macrophages. RIA expression pattern was used for a new model of interaction between L. corymbifera and macrophages.
Collapse
Affiliation(s)
- Felicia Adelina Stanford
- Jena Microbial Resource Collection, Leibniz Institute for Natural Product Research, and Infection Biology—Hans Knöll Institute (HKI), 07745 Jena, Germany; (F.A.S.); (N.M.); (M.I.A.H.)
- Institute of Microbiology, Friedrich Schiller University Jena, 07743 Jena, Germany;
| | - Nina Matthies
- Jena Microbial Resource Collection, Leibniz Institute for Natural Product Research, and Infection Biology—Hans Knöll Institute (HKI), 07745 Jena, Germany; (F.A.S.); (N.M.); (M.I.A.H.)
- Institute of Microbiology, Friedrich Schiller University Jena, 07743 Jena, Germany;
| | - Zoltán Cseresnyés
- Applied Systems Biology, Leibniz Institute for Natural Product Research, and Infection Biology—Hans Knöll Institute, 12622 Jena, Germany;
| | - Marc Thilo Figge
- Institute of Microbiology, Friedrich Schiller University Jena, 07743 Jena, Germany;
- Applied Systems Biology, Leibniz Institute for Natural Product Research, and Infection Biology—Hans Knöll Institute, 12622 Jena, Germany;
| | - Mohamed I. Abdelwahab Hassan
- Jena Microbial Resource Collection, Leibniz Institute for Natural Product Research, and Infection Biology—Hans Knöll Institute (HKI), 07745 Jena, Germany; (F.A.S.); (N.M.); (M.I.A.H.)
- Institute of Microbiology, Friedrich Schiller University Jena, 07743 Jena, Germany;
- National Research Centre, Pests & Plant Protection Department, 33rd El Buhouth St., Dokki, Giza 12622, Egypt
| | - Kerstin Voigt
- Jena Microbial Resource Collection, Leibniz Institute for Natural Product Research, and Infection Biology—Hans Knöll Institute (HKI), 07745 Jena, Germany; (F.A.S.); (N.M.); (M.I.A.H.)
- Institute of Microbiology, Friedrich Schiller University Jena, 07743 Jena, Germany;
- Correspondence: or ; Tel.: +49-3641-532-1395
| |
Collapse
|
30
|
Borman AM, Fraser M, Patterson Z, Palmer MD, Johnson EM. In Vitro Antifungal Drug Resistance Profiles of Clinically Relevant Members of the Mucorales (Mucoromycota) Especially with the Newer Triazoles. J Fungi (Basel) 2021; 7:271. [PMID: 33918216 PMCID: PMC8065934 DOI: 10.3390/jof7040271] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 03/25/2021] [Accepted: 03/31/2021] [Indexed: 12/23/2022] Open
Abstract
Mucoromycoses (infections caused by members of the order Mucorales, phylum Mucoromycota [ex-Zygomycota]) are highly destructive, rapidly progressive infections, with dire prognoses especially when they occur in immunocompromised hosts. Current treatment guidelines recommend liposomal formulations of amphotericin B with adjunctive surgery as first line therapy, with the newer triazoles posaconazole or isavuconazole as alternative treatments, or as salvage therapy. Among the many organisms belonging to this order, a limited number of species in the genera Rhizopus, Mucor, Lichtheimia and Rhizomucor are responsible for most cases of human infection. Here, we present the minimum inhibitory concentration data (MICs) for amphotericin B, posaconazole, isavuconazole, itraconazole and voriconazole with a panel of over 300 isolates of the five most common agents of human infection (Lichtheimia corymbifera, Rhizopus arrhizus, R. microsporus, Rhizomucor pusillus and Mucor spp.) determined using the CLSI broth microdilution method. In agreement with previous studies, the most active antifungal drug for all Mucorales was amphotericin B, with MICs within the range that would predict susceptibility with Aspergillus fumigatus. Conversely, MICs for voriconazole against all species tested were high, and above the range associated with clinical efficacy with A. fumigatus. Interestingly, whilst isavuconazole and posaconazole MIC distributions indicated in vitro activity against some members of the Mucorales, activity was species-dependent for both agents. These data underscore the importance of accurate identification of the causative agents of mucoromycosis, coupled with antifungal susceptibility testing of individual isolates, in determining the optimal treatment of infections caused by these aggressive opportunistic human fungal pathogens.
Collapse
Affiliation(s)
- Andrew M. Borman
- UK National Mycology Reference Laboratory, Public Health England, Science Quarter, Southmead Hospital, Bristol BS10 5NB, UK; (M.F.); (Z.P.); (M.D.P.)
- Medical Research Council Centre for Medical Mycology (MRC CMM), University of Exeter, Exeter EX4 4QD, UK
| | - Mark Fraser
- UK National Mycology Reference Laboratory, Public Health England, Science Quarter, Southmead Hospital, Bristol BS10 5NB, UK; (M.F.); (Z.P.); (M.D.P.)
| | - Zoe Patterson
- UK National Mycology Reference Laboratory, Public Health England, Science Quarter, Southmead Hospital, Bristol BS10 5NB, UK; (M.F.); (Z.P.); (M.D.P.)
| | - Michael D. Palmer
- UK National Mycology Reference Laboratory, Public Health England, Science Quarter, Southmead Hospital, Bristol BS10 5NB, UK; (M.F.); (Z.P.); (M.D.P.)
| | - Elizabeth M. Johnson
- UK National Mycology Reference Laboratory, Public Health England, Science Quarter, Southmead Hospital, Bristol BS10 5NB, UK; (M.F.); (Z.P.); (M.D.P.)
- Medical Research Council Centre for Medical Mycology (MRC CMM), University of Exeter, Exeter EX4 4QD, UK
| |
Collapse
|
31
|
Neofytos D, Garcia-Vidal C, Lamoth F, Lichtenstern C, Perrella A, Vehreschild JJ. Invasive aspergillosis in solid organ transplant patients: diagnosis, prophylaxis, treatment, and assessment of response. BMC Infect Dis 2021; 21:296. [PMID: 33761875 PMCID: PMC7989085 DOI: 10.1186/s12879-021-05958-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Accepted: 03/04/2021] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Invasive aspergillosis (IA) is a rare complication in solid organ transplant (SOT) recipients. Although IA has significant implications on graft and patient survival, data on diagnosis and management of this infection in SOT recipients are still limited. METHODS Discussion of current practices and limitations in the diagnosis, prophylaxis, and treatment of IA and proposal of means of assessing treatment response in SOT recipients. RESULTS Liver, lung, heart or kidney transplant recipients have common as well as different risk factors to the development of IA, thus each category needs a separate evaluation. Diagnosis of IA in SOT recipients requires a high degree of awareness, because established diagnostic tools may not provide the same sensitivity and specificity observed in the neutropenic population. IA treatment relies primarily on mold-active triazoles, but potential interactions with immunosuppressants and other concomitant therapies need special attention. CONCLUSIONS Criteria to assess response have not been sufficiently evaluated in the SOT population and CT lesion dynamics, and serologic markers may be influenced by the underlying disease and type and severity of immunosuppression. There is a need for well-orchestrated efforts to study IA diagnosis and management in SOT recipients and to develop comprehensive guidelines for this population.
Collapse
Affiliation(s)
- Dionysios Neofytos
- Service des Maladies Infectieuses, Hôpitaux Universitaires de Genève, Rue Gabrielle-Perret-Gentil 4, Geneva, Switzerland.
| | - Carolina Garcia-Vidal
- Servicio de Enfermedades Infecciosas, Hospital Clínic de Barcelona-IDIBAPS, Universitat de Barcelona, FungiCLINIC Research group (AGAUR), Barcelona, Spain
| | - Frédéric Lamoth
- Infectious Diseases Service, Department of Medicine, Lausanne University Hospital, 1011, Lausanne, Switzerland
- Department of Laboratories, Institute of Microbiology, Lausanne University Hospital, Lausanne, Switzerland
| | - Christoph Lichtenstern
- Department of Anaesthesiology, Heidelberg University Hospital, Im Neuenheimer Feld 110, Heidelberg, Germany
| | - Alessandro Perrella
- VII Department of Infectious Disease and Immunology, Hospital D. Cotugno, Naples, Italy
- CLSE-Liver Transplant Unit, Hospital A. Cardarelli, Naples, Italy
| | - Jörg Janne Vehreschild
- Medical Department II, Hematology and Oncology, University Hospital of Frankfurt, Frankfurt, Germany
- Department I for Internal Medicine, University Hospital of Cologne, Cologne, Germany
- German Centre for Infection Research, partner site Bonn-Cologne, University of Cologne, Cologne, Germany
| |
Collapse
|
32
|
Breakthrough Mucormycosis Developing on Mucorales-Active Antifungals Portrays a Poor Prognosis in Patients with Hematologic Cancer. J Fungi (Basel) 2021; 7:jof7030217. [PMID: 33802827 PMCID: PMC8002622 DOI: 10.3390/jof7030217] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 03/12/2021] [Accepted: 03/15/2021] [Indexed: 01/15/2023] Open
Abstract
Although breakthrough mucormycosis (BT-MCR) is known to develop on mold-active antifungals without Mucorales activity, it can also occur while on Mucorales-active antifungals. Herein, we retrospectively compared the characteristics and outcomes of patients with hematologic malignancies (HMs) or hematopoietic stem cell transplant (HSCT) who developed BT-MCR on mold-active antifungals with or without Mucorales activity. Of the patients developing BT-MCR, 16 were on Mucorales-active antifungals (9 isavuconazole, 6 posaconazole, 1 amphotericin B), and 87 were on other mold-active agents (52 voriconazole, 22 echinocandins, 8 itraconazole, 5 echinocandin + voriconazole). Both groups were largely comparable in clinical characteristics. Patients developing BT-MCR while on Mucorales-active antifungals had higher 42-day mortality, from either symptom onset (63% versus 25%, p = 0.006) or treatment initiation (69% versus 39%, p = 0.028). In multivariate Cox regression analysis, exposure to Mucorales-active antifungals prior to BT-MCR had a hazard ratio of 2.40 (p = 0.015) for 42-day mortality from treatment initiation and 4.63 (p < 0.001) for 42-day mortality from symptom onset. Intensive care unit (ICU) admission and APACHE II score at diagnosis, non-recovered severe neutropenia, active HM, and amphotericin B/caspofungin combination treatment were additional independent predictors of 42-day mortality. In summary, BT-MCR on Mucorales-active antifungals portrays poor prognosis in HM/HSCT patients. Moreover, improvements in early diagnosis and treatment are urgently needed in these patients.
Collapse
|
33
|
Postextraction Mucormycosis in Immunocompromised-Patient Management and Review of Literature. J Oral Maxillofac Surg 2021; 79:1482-1491. [PMID: 33617788 DOI: 10.1016/j.joms.2021.01.019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 01/16/2021] [Accepted: 01/16/2021] [Indexed: 11/23/2022]
Abstract
PURPOSE Mucormycosis is an uncommon aggressive, opportunistic infection that can produce widespread orofacial tissue necrosis. This primarily affects immunocompromised individuals. It is the deadliest and most rapidly progressing type of human-affecting fungal infection. The aim of the study is to investigate the diagnostic criteria and treatment approach in 10 cases of mucormycosis in the author's institute from 2008 to 2019. Moreover, a review of the English literature presents all cases of mucormycosis after tooth extraction. MATERIALS AND METHODS Ten patients at our institute have been treated. They were evaluated and discussed as per their diagnostic criteria, surgical treatment, and mortality rates. RESULTS All 10 immunocompromised patients had a primary hematologic malignancy. The criteria for a positive diagnosis were clinical symptoms and a biopsy for microbiological culture and histologic analysis. Each patient was subjected to vigorous surgical resection and underwent antifungal treatment. Seven patients died because of their main disease. Owing to the unregulated spread of mucormycosis, 2 patients died. Four cases were diagnosed after maxillary tooth extraction. CONCLUSIONS The most important factor for patient survival tends to be the management of the underlying disease with early detection and active surgical and antifungal action. Four of 10 cases diagnosed with mucormycosis appeared after tooth extraction, a relatively high number compared with the literature. Therefore, it is the dental profession's obligation to be familiar with the possibility of the potentially severe and possibly fatal complication.
Collapse
|
34
|
Gangneux JP, Padoin C, Michallet M, Saillio E, Kumichel A, Peffault de La Tour R, Ceballos P, Gastinne T, Pigneux A. Outcomes of Antifungal Prophylaxis in High-Risk Haematological Patients (AML under Intensive Chemotherapy): The SAPHIR Prospective Multicentre Study. J Fungi (Basel) 2020; 6:jof6040281. [PMID: 33198192 PMCID: PMC7712136 DOI: 10.3390/jof6040281] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 11/09/2020] [Accepted: 11/10/2020] [Indexed: 12/15/2022] Open
Abstract
Antifungal prophylaxis (AFP) is recommended by international guidelines for patients with acute myeloid leukaemia (AML) undergoing induction chemotherapy and allogeneic hematopoietic cell transplantation. Nonetheless, treatment of breakthrough fungal infections remains challenging. This observational, prospective, multicentre, non-comparative study of patients undergoing myelosuppressive and intensive chemotherapy for AML who are at high-risk of invasive fungal diseases (IFDs), describes AFP management and outcomes for 404 patients (65.6% newly diagnosed and 73.3% chemotherapy naïve). Ongoing chemotherapy started 1.0 ± 4.5 days before inclusion and represented induction therapy for 79% of participants. In 92.3% of patients, posaconazole was initially prescribed, and 8.2% of all patients underwent at least one treatment change after 17 ± 24 days, mainly due to medical conditions influencing AFP absorption (65%). The mean AFP period was 24 ± 32 days, 66.8% stopped their prophylaxis after the high-risk period and 31.2% switched to a non-prophylactic treatment (2/3 empirical, 1/3 pre-emptive/curative). Overall, 9/404 patients (2.2%) were diagnosed with probable or proven IFDs. During the follow-up, 94.3% showed no signs of infection. Altogether, 20 patients (5%) died, and three deaths (0.7%) were IFD-related. In conclusion, AFP was frequently prescribed and well tolerated by these AML patients, breakthrough infections incidence and IFD mortality were low and very few treatment changes were required.
Collapse
Affiliation(s)
- Jean-Pierre Gangneux
- Mycology Department, Centre Hospitalier Universitaire de Rennes, University Rennes, INSERM, Irset (Institut de Recherche en Santé, Environnement et Travail), UMR S_1085, 35000 Rennes, France
- Correspondence: ; Tel.: +33-299-283-731
| | - Christophe Padoin
- Pharmacy Department, CHU Martinique Site P. Zobda Quitman, 97261 Fort de France, Martinique, France;
| | - Mauricette Michallet
- Clinical Haematology Department, Centre Léon Bérard (Anticancer Center), 28 Rue Laennec, 69373 Lyon, France;
| | - Emeline Saillio
- Department of Medical Affairs, MSD France, 10-12 cours Michelet, 92800 Puteaux, France;
| | - Alexandra Kumichel
- Scientific Department, ClinSearch, 110 Avenue Pierre Brossolette, 92240 Malakoff, France;
| | - Régis Peffault de La Tour
- Haematology-Bone Marrow Transplant Department, Saint-Louis Hospital APHP, 1 Avenue Claude-Vellefaux, 75010 Paris, France;
| | - Patrice Ceballos
- Clinical Haematology Department, CHRU Lapeyronie, 371 Avenue Doyen Gaston Giraud, 34295 Montpellier, France;
| | - Thomas Gastinne
- Clinical Haematology Department, CHU Nantes, 1 Place Alexis-Ricordeau, 44093 Nantes, France;
| | - Arnaud Pigneux
- Blood Diseases Department, Hospital Group Haut Leveque, Avenue de Magellan, 33604 Pessac, France;
| |
Collapse
|
35
|
Skiada A, Pavleas I, Drogari-Apiranthitou M. Epidemiology and Diagnosis of Mucormycosis: An Update. J Fungi (Basel) 2020; 6:jof6040265. [PMID: 33147877 PMCID: PMC7711598 DOI: 10.3390/jof6040265] [Citation(s) in RCA: 295] [Impact Index Per Article: 73.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 10/30/2020] [Accepted: 11/01/2020] [Indexed: 12/17/2022] Open
Abstract
Mucormycosis is an angioinvasive fungal infection, due to fungi of the order Mucorales. Its incidence cannot be measured exactly, since there are few population-based studies, but multiple studies have shown that it is increasing. The prevalence of mucormycosis in India is about 80 times the prevalence in developed countries, being approximately 0.14 cases per 1000 population. Diabetes mellitus is the main underlying disease globally, especially in low and middle-income countries. In developed countries the most common underlying diseases are hematological malignancies and transplantation. Τhe epidemiology of mucormycosis is evolving as new immunomodulating agents are used in the treatment of cancer and autoimmune diseases, and as the modern diagnostic tools lead to the identification of previously uncommon genera/species such as Apophysomyces or Saksenaea complex. In addition, new risk factors are reported from Asia, including post-pulmonary tuberculosis and chronic kidney disease. New emerging species include Rhizopus homothallicus, Thamnostylum lucknowense, Mucor irregularis and Saksenaea erythrospora. Diagnosis of mucormycosis remains challenging. Clinical approach to diagnosis has a low sensitivity and specificity, it helps however in raising suspicion and prompting the initiation of laboratory testing. Histopathology, direct examination and culture remain essential tools, although the molecular methods are improving. The internal transcribed spacer (ITS) region is the most widely sequenced DNA region for fungi and it is recommended as a first-line method for species identification of Mucorales. New molecular platforms are being investigated and new fungal genetic targets are being explored. Molecular-based methods have gained acceptance for confirmation of the infection when applied on tissues. Methods on the detection of Mucorales DNA in blood have shown promising results for earlier and rapid diagnosis and could be used as screening tests in high-risk patients, but have to be validated in clinical studies. More, much needed, rapid methods that do not require invasive procedures, such as serology-based point-of-care, or metabolomics-based breath tests, are being developed and hopefully will be evaluated in the near future.
Collapse
Affiliation(s)
- Anna Skiada
- First Department of Medicine, Laiko Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece
- Correspondence: ; Tel.: +30-2107-462-607
| | | | - Maria Drogari-Apiranthitou
- Fourth Department of Internal Medicine, General University Hospital “Attikon”, National and Kapodistrian University of Athens, 12462 Athens, Greece;
| |
Collapse
|
36
|
Wong TY, Loo YS, Veettil SK, Wong PS, Divya G, Ching SM, Menon RK. Efficacy and safety of posaconazole for the prevention of invasive fungal infections in immunocompromised patients: a systematic review with meta-analysis and trial sequential analysis. Sci Rep 2020; 10:14575. [PMID: 32884060 PMCID: PMC7471265 DOI: 10.1038/s41598-020-71571-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Accepted: 08/17/2020] [Indexed: 12/13/2022] Open
Abstract
Invasive fungal infections are a potentially life-threatening complication in immunocompromised patients. The aim of this study was to assess the efficacy and safety of posaconazole as compared with other antifungal agents for preventing invasive fungal infections in immunocompromised patients. Embase, CENTRAL, and MEDLINE were searched for randomized conweekmonthtrolled trials (RCTs) up to June 2020. A systematic review with meta-analysis of RCTs was performed using random-effects model. Trial sequential analysis (TSA) was conducted for the primary outcome to assess random errors. A total of five RCTs with 1,617 participants were included. Posaconazole prophylaxis was associated with a significantly lower risk of IFIs (RR, 0.43 [95% CI 0.28 to 0.66, p = 0.0001]) as compared to other antifungal agents. No heterogeneity was identified between studies (I2 = 0%). No significant associations were observed for the secondary outcomes measured, including risk reduction of invasive aspergillosis and candidiasis, clinical failure, all-cause mortality, and treatment-related adverse events, except for infection-related mortality (RR, 0.31 [95% CI 0.15 to 0.64, p = 0.0001]). Subgroup analysis favoured posaconazole over fluconazole for the prevention of IFIs (RR, 0.44 [95% CI 0.28 to 0.70, p = 0.0004]). TSA confirmed the prophylactic benefit of posaconazole against IFIs. Posaconazole is effective in preventing IFIs among immunocompromised patients, particularly those with hematologic malignancies and recipients of allogenic hematopoietic stem cell transplantation.
Collapse
Affiliation(s)
- Tse Yee Wong
- School of Pharmacy, International Medical University, Kuala Lumpur, Malaysia
| | - Yee Shen Loo
- School of Pharmacy, International Medical University, Kuala Lumpur, Malaysia
| | - Sajesh Kalkandi Veettil
- Department of Pharmacy Practice, School of Pharmacy, International Medical University, Kuala Lumpur, Malaysia.
| | - Pei Se Wong
- Department of Pharmacy Practice, School of Pharmacy, International Medical University, Kuala Lumpur, Malaysia
| | - Gopinath Divya
- Oral Diagnostic and Surgical Sciences, School of Dentistry, International Medical University, Kuala Lumpur, Malaysia
| | - Siew Mooi Ching
- Department of Family Medicine, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Malaysia
| | - Rohit Kunnath Menon
- Division of Clinical Dentistry, School of Dentistry, International Medical University, Kuala Lumpur, Malaysia.
| |
Collapse
|
37
|
Cerebral and pulmonary aspergillosis, treatment and diagnostic challenges of mixed breakthrough invasive fungal infections: case report study. BMC Infect Dis 2020; 20:535. [PMID: 32703183 PMCID: PMC7376672 DOI: 10.1186/s12879-020-05162-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Accepted: 06/17/2020] [Indexed: 02/08/2023] Open
Abstract
Background Breakthrough invasive fungal infections (bIFIs) are an area of concern in the scarcity of new antifungals. The mixed form of bIFIs is a rare phenomenon but could be potentially a troublesome challenge when caused by azole-resistant strains or non-Aspergillus fumigatus. To raise awareness and emphasize diagnostic challenges, we present a case of mixed bIFIs in a child with acute lymphoblastic leukemia. Case presentation A newly diagnosed 18-month-old boy with acute lymphoblastic leukemia was complicated with prolonged severe neutropenia after induction chemotherapy. He experienced repeated episodes of fever due to extended-spectrum beta-lactamase-producing Escherichia coli bloodstream infection and pulmonary invasive fungal infection with Aspergillus fumigatus (early-type bIFIs) while receiving antifungal prophylaxis. Shortly after pulmonary involvement, his condition aggravated by abnormal focal movement, loss of consciousness and seizure. Cerebral aspergillosis with Aspergillus niger diagnosed after brain tissue biopsy. The patient finally died despite 108-day antifungal therapy. Conclusions Mixed bIFIs is a rare condition with high morbidity and mortality in the patients receiving immunosuppressants for hematological malignancies. This case highlights the clinical importance of Aspergillus identification at the species level in invasive fungal infections with multiple site involvement in the patients on antifungal prophylaxis.
Collapse
|
38
|
Zaniol E, Daprà V, Filomena R, Alliaudi C, Calvi C, Montanari P, Galliano I, Bergallo M. Real time qPCR TaqMan method for detection of Fusarium solani. MINERVA BIOTECNOL 2020. [DOI: 10.23736/s1120-4826.20.02621-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
39
|
Signorelli J, Lei M, Lam J, Jalbut M, Amrein PC, Fathi AT, Hobbs G, Hock H, McAfee SL, Letourneau AR, Narayan R, Brunner A. Incidence of Invasive Fungal Infections in Acute Myeloid Leukemia Without Antifungal Prophylaxis. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2020; 20:e883-e889. [PMID: 32917574 DOI: 10.1016/j.clml.2020.06.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 06/12/2020] [Accepted: 06/16/2020] [Indexed: 11/29/2022]
Abstract
BACKGROUND Antifungal prophylaxis during induction for acute myeloid leukemia (AML) varies according to local rates of invasive fungal infections (IFIs). We evaluated fluconazole prophylaxis and no antifungal prophylaxis, as a natural interrupted time-series study to assess survival and infection complications. PATIENTS AND METHODS We identified patients with AML ≥ 18 years old undergoing induction chemotherapy during 2 time periods: period 1, fluconazole prophylaxis from August 1, 2013 to September 30, 2015, and period 2, no prophylaxis from October 1, 2015 to December 31, 2017. The primary outcome was incidence of proven or probable IFI. Secondary outcomes included types of IFIs and 60-day overall survival (OS). IFI was defined by the 2002 European Organization for Research and Treatment of Cancer/Mycoses Study Group Consensus criteria. RESULTS One hundred forty-four patients received induction chemotherapy over the 2 time periods. In the prophylaxis versus no-prophylaxis groups, the rate of proven or probable IFIs was 4 (5%) of 87 versus 12 (21%) of 57 (P = .01). The total number of proven IFIs was 3 (3%) of 87 versus 4 (7%) of 57 (P = .44), whereas probable IFIs were 1 (1%) of 87 versus 8 (14%) of 57 (P < .01). No difference was observed in fungemia. Incidence of IFIs was too low to detect resistance patterns. OS at 60 days was improved in with fluconazole prophylaxis compared with no prophylaxis (hazard ratio, 0.329; 95% confidence interval, 0.12-0.89; P = .028). CONCLUSION Observed rates of proven or probable IFI were lower in the fluconazole prophylaxis group versus the no-prophylaxis group. Sixty-day OS was higher with fluconazole prophylaxis. Further study is required to evaluate how fluconazole may impart the differences in survival seen in this analysis.
Collapse
Affiliation(s)
| | - Matthew Lei
- Department of Pharmacy, Massachusetts General Hospital, Boston, MA
| | - Jenna Lam
- Department of Pharmacy, Massachusetts General Hospital, Boston, MA
| | - Marla Jalbut
- Division of Hematology/Oncology, Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Philip C Amrein
- Division of Hematology/Oncology, Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Amir T Fathi
- Division of Hematology/Oncology, Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Gabriela Hobbs
- Division of Hematology/Oncology, Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Hanno Hock
- Division of Hematology/Oncology, Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Steven Lawrence McAfee
- Division of Hematology/Oncology, Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Alyssa R Letourneau
- Division of Infectious Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Rupa Narayan
- Division of Hematology/Oncology, Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Andrew Brunner
- Division of Hematology/Oncology, Department of Medicine, Massachusetts General Hospital, Boston, MA
| |
Collapse
|
40
|
Posteraro B, De Carolis E, Criscuolo M, Ballanti S, De Angelis G, Del Principe MI, Delia M, Fracchiolla N, Marchesi F, Nadali G, Picardi M, Piccioni AL, Verga L, Candoni A, Busca A, Sanguinetti M, Pagano L. Candidaemia in haematological malignancy patients from a SEIFEM study: Epidemiological patterns according to antifungal prophylaxis. Mycoses 2020; 63:900-910. [PMID: 32531854 DOI: 10.1111/myc.13130] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 06/03/2020] [Accepted: 06/04/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND Candidaemia is an important infectious complication for haematological malignancy patients. Antifungal prophylaxis reduces the incidence of candidaemia but may be associated with breakthrough candidaemia. OBJECTIVE To analyse the Candida species' distribution and relative antifungal susceptibility profiles of candidaemia episodes in relation to the use of antifungal prophylaxis among Italian SEIFEM haematology centres. METHODOLOGY This multicentre retrospective observational SEIFEM study included 133 single-species candidaemia episodes of haematological malignancy patients for whom antifungal susceptibility testing results of blood Candida isolates were available between 2011 and 2015. Each participating centre provided both clinical and microbiological data. RESULTS Non-Candida albicans Candida (NCAC) species were the mostly isolated species (89, 66.9%), which accounted for C parapsilosis (35, 26.3%), C glabrata (16, 12.0%), C krusei (14, 10.5%), C tropicalis (13, 9.8%) and uncommon species (11, 8.3%). C albicans caused the remaining 44 (33.1%) episodes. Excluding 2 C albicans isolates, 23 of 25 fluconazole-resistant isolates were NCAC species (14 C krusei, 6 C glabrata, 2 C parapsilosis and 1 C tropicalis). Fifty-six (42.1%) of 133 patients developed breakthrough candidaemia. Systemic antifungal prophylaxis consisted of azoles, especially fluconazole and posaconazole, in 50 (89.3%) of 56 patients in whom a breakthrough candidaemia occurred. Interestingly, all these patients tended to develop a C krusei infection (10/56, P = .02) or a fluconazole-resistant isolate's infection (14/50, P = .04) compared to patients (4/77 and 10/77, respectively) who did not have a breakthrough candidaemia. CONCLUSIONS Optimisation of prophylactic strategies is necessary to limit the occurrence of breakthrough candidaemia and, importantly, the emergence of fluconazole-resistant NCAC isolates' infections in haematological malignancy patients.
Collapse
Affiliation(s)
- Brunella Posteraro
- Dipartimento di Scienze Gastroenterologiche, Endocrino-Metaboliche e Nefro-Urologiche, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy.,Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie, Università Cattolica del Sacro Cuore, Roma, Italy
| | - Elena De Carolis
- Dipartimento di Scienze di Laboratorio e Infettivologiche, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy
| | - Marianna Criscuolo
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy
| | - Stelvio Ballanti
- Dipartimento di Ematologia, Ospedale Santa Maria della Misericordia, Università di Perugia, Perugia, Italy
| | - Giulia De Angelis
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie, Università Cattolica del Sacro Cuore, Roma, Italy
| | | | - Mario Delia
- Dipartimento dell'Emergenza e dei Trapianti di Organo, Azienda Ospedaliero-Universitaria Policlinico di Bari, Bari, Italy
| | - Nicola Fracchiolla
- Unità di Ematologia, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Francesco Marchesi
- Unità di Ematologia e Trapianti, Istituto Nazionale Tumori Regina Elena IRCCS, Roma, Italy
| | - Gianpaolo Nadali
- Unità di Ematologia, Dipartimento di Medicina, Università di Verona, Verona, Italy
| | - Marco Picardi
- Dipartimento di Scienze Biomediche Avanzate, Azienda Ospedaliera Universitaria Federico II di Napoli, Napoli, Italy
| | - Anna Lina Piccioni
- Dipartimento di Ematologia, Azienda Ospedaliera San Giovanni Addolorata, Roma, Italy
| | - Luisa Verga
- Ematologia Adulti e CTA, Ospedale San Gerardo, Monza, Italy
| | - Anna Candoni
- Centro Trapianti e Terapie Cellulari, Azienda Sanitaria Universitaria Integrata di Udine, Udine, Italy
| | - Alessandro Busca
- Centro Trapianti di Midollo, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza, Torino, Italy
| | - Maurizio Sanguinetti
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie, Università Cattolica del Sacro Cuore, Roma, Italy.,Dipartimento di Scienze di Laboratorio e Infettivologiche, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy
| | - Livio Pagano
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy.,Dipartimento di Scienze Radiologiche ed Ematologiche, Università Cattolica del Sacro Cuore, Roma, Italy
| | | |
Collapse
|
41
|
Ruhnke M, Cornely OA, Schmidt-Hieber M, Alakel N, Boell B, Buchheidt D, Christopeit M, Hasenkamp J, Heinz WJ, Hentrich M, Karthaus M, Koldehoff M, Maschmeyer G, Panse J, Penack O, Schleicher J, Teschner D, Ullmann AJ, Vehreschild M, von Lilienfeld-Toal M, Weissinger F, Schwartz S. Treatment of invasive fungal diseases in cancer patients-Revised 2019 Recommendations of the Infectious Diseases Working Party (AGIHO) of the German Society of Hematology and Oncology (DGHO). Mycoses 2020; 63:653-682. [PMID: 32236989 DOI: 10.1111/myc.13082] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 03/05/2020] [Accepted: 03/10/2020] [Indexed: 12/15/2022]
Abstract
BACKGROUND Invasive fungal diseases remain a major cause of morbidity and mortality in cancer patients undergoing intensive cytotoxic therapy. The choice of the most appropriate antifungal treatment (AFT) depends on the fungal species suspected or identified, the patient's risk factors (eg length and depth of granulocytopenia) and the expected side effects. OBJECTIVES Since the last edition of recommendations for 'Treatment of invasive fungal infections in cancer patients' of the Infectious Diseases Working Party (AGIHO) of the German Society of Hematology and Medical Oncology (DGHO) in 2013, treatment strategies were gradually moving away from solely empirical therapy of presumed or possible invasive fungal diseases (IFDs) towards pre-emptive therapy of probable IFD. METHODS The guideline was prepared by German clinical experts for infections in cancer patients in a stepwise consensus process. MEDLINE was systematically searched for English-language publications from January 1975 up to September 2019 using the key terms such as 'invasive fungal infection' and/or 'invasive fungal disease' and at least one of the following: antifungal agents, cancer, haematological malignancy, antifungal therapy, neutropenia, granulocytopenia, mycoses, aspergillosis, candidosis and mucormycosis. RESULTS AFT of IFDs in cancer patients may include not only antifungal agents but also non-pharmacologic treatment. In addition, the armamentarium of antifungals for treatment of IFDs has been broadened (eg licensing of isavuconazole). Additional antifungals are currently under investigation or in clinical trials. CONCLUSIONS Here, updated recommendations for the treatment of proven or probable IFDs are given. All recommendations including the levels of evidence are summarised in tables to give the reader rapid access to key information.
Collapse
Affiliation(s)
- Markus Ruhnke
- Division of Haematology, Oncology and Palliative Care, Department of Internal Medicine, Evangelisches Klinikum Bethel, Bielefeld, Germany
| | - Oliver A Cornely
- Department I of Internal Medicine, Faculty of Medicine, University of Cologne, Cologne, Germany.,ECMM Excellence Centre of Medical Mycology, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany.,Clinical Trials Centre Cologne (ZKS Köln), University of Cologne, Cologne, Germany
| | | | - Nael Alakel
- Department I of Internal Medicine, Haematology and Oncology, University Hospital Dresden, Dresden, Germany
| | - Boris Boell
- Department I of Internal Medicine, Faculty of Medicine, University of Cologne, Cologne, Germany
| | - Dieter Buchheidt
- Department of Hematology and Oncology, Mannheim University Hospital, Heidelberg University, Mannheim, Germany
| | - Maximilian Christopeit
- Department of Stem Cell Transplantation & Oncology, University Medical Center Eppendorf, Hamburg, Germany
| | - Justin Hasenkamp
- Clinic for Haematology and Medical Oncology with Department for Stem Cell Transplantation, University Medicine Göttingen, Göttingen, Germany
| | - Werner J Heinz
- Schwerpunkt Infektiologie, Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, Würzburg, Germany
| | - Marcus Hentrich
- Hämatologie und Internistische Onkologie, Innere Medizin III, Rotkreuzklinikum München, München, Germany
| | - Meinolf Karthaus
- Department of Haematology & Oncology, Municipal Hospital Neuperlach, München, Germany
| | - Michael Koldehoff
- Klinik für Knochenmarktransplantation, Westdeutsches Tumorzentrum Essen, Universitätsklinikum Essen (AöR), Essen, Germany
| | - Georg Maschmeyer
- Department of Hematology, Onclogy and Palliative Care, Klinikum Ernst von Bergmann, Potsdam, Germany
| | - Jens Panse
- Klinik für Onkologie, Hämatologie und Stammzelltransplantation, Universitätsklinikum Aachen, Aachen, Germany
| | - Olaf Penack
- Division of Haematology & Oncology, Department of Internal Medicine, Charité University Medicine, Campus Rudolf Virchow, Berlin, Germany
| | - Jan Schleicher
- Klinik für Hämatologie Onkologie und Palliativmedizin, Katharinenhospital, Stuttgart, Germany
| | - Daniel Teschner
- III. Medizinische Klinik und Poliklinik, Universitätsmedizin der Johannes Gutenberg-Universität Mainz, Mainz, Germany
| | - Andrew John Ullmann
- Department of Internal Medicine II, Julius Maximilians University, Würzburg, Germany
| | - Maria Vehreschild
- Department I of Internal Medicine, Faculty of Medicine, University of Cologne, Cologne, Germany.,ECMM Excellence Centre of Medical Mycology, Cologne, Germany.,Zentrum für Innere Medizin, Infektiologie, Goethe Universität Frankfurt, Frankfurt am Main, Deutschland.,Deutsches Zentrum für Infektionsforschung (DZIF), Standort Bonn-Köln, Deutschland
| | - Marie von Lilienfeld-Toal
- Klinik für Innere Medizin II, Abteilung für Hämatologie und Internistische Onkologie, Universitätsklinikum Jena, Jena, Germany
| | - Florian Weissinger
- Division of Haematology, Oncology and Palliative Care, Department of Internal Medicine, Evangelisches Klinikum Bethel, Bielefeld, Germany
| | - Stefan Schwartz
- Division of Haematology & Oncology, Department of Internal Medicine, Charité University Medicine, Campus Benjamin Franklin, Berlin, Germany
| |
Collapse
|
42
|
|
43
|
Stohs E, Zimmer A. An Approach to Suspected Invasive Fungal Infection in Patients with Hematologic Malignancy and HCT Recipients with Persistent Neutropenic Fever Despite Mold-Active Prophylaxis. CURRENT FUNGAL INFECTION REPORTS 2020. [DOI: 10.1007/s12281-020-00375-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
44
|
Breakthrough Invasive Fungal Infections in Patients with Acute Myeloid Leukemia. Mycopathologia 2020; 185:299-306. [PMID: 31939052 DOI: 10.1007/s11046-019-00418-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 12/10/2019] [Indexed: 01/01/2023]
Abstract
OBJECTIVES We sought to determine the occurrence, risk factors, effect of antifungal prophylaxis, and outcomes of invasive fungal infections (IFIs) in patients with acute myeloid leukemia (AML). PATIENTS AND METHODS We performed a retrospective analysis of all adult patients admitted to the University of Michigan Health System for AML over a 3-year period from 2010 to 2013. We determined comorbidities, hematopoietic cell transplant (HCT) status, antifungal prophylaxis, proven and probable IFI, and outcomes at 12 weeks after initiation of appropriate antifungal therapy. RESULTS Of 333 patients in our cohort, 116 of whom had received a HCT, 98 (29%) developed an IFI. Of the 30 (9%) patients who had a proven or probable IFI, 18 had breakthrough infection while on micafungin (n = 5), voriconazole (n = 4), posaconazole (n = 5), or fluconazole (n = 4). Breakthrough IFIs were due to Aspergillus species (n = 11), other molds (n = 4), and Candida species (n = 3). Factors associated with breakthrough IFI were prolonged severe neutropenia (p = .05) and having received tacrolimus (p = .04). Antifungal therapy was successful in 7 of the 18 (39%) patients with breakthrough IFI and 8 of the 12 (67%) patients with non-breakthrough IFI, p = .13. Mortality at 12 weeks was 27%, 5 with breakthrough IFI and 3 with non-breakthrough IFI and was associated with prolonged severe neutropenia, p = .04. CONCLUSIONS Patients with AML remain at risk for IFI despite the use of several different antifungal agents for prophylaxis. Mortality remains high in patients with AML who develop IFI.
Collapse
|
45
|
Lee HJ, Cho SY, Lee DG, Park C, Chun HS, Park YJ. Characteristics and risk factors for mortality of invasive non-Aspergillus mould infections in patients with haematologic diseases: A single-centre 7-year cohort study. Mycoses 2019; 63:257-264. [PMID: 31762083 PMCID: PMC7065074 DOI: 10.1111/myc.13038] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 11/14/2019] [Accepted: 11/19/2019] [Indexed: 12/13/2022]
Abstract
Since mould-active azole prophylaxis has become a standard approach for patients with high-risk haematologic diseases, the epidemiology of invasive fungal infections (IFIs) has shifted towards non-Aspergillus moulds. It was aimed to identify the epidemiology and characteristics of non-Aspergillus invasive mould infections (NAIMIs). Proven/probable NAIMIs developed in patients with haematologic diseases were reviewed from January 2011 to August 2018 at Catholic Hematology hospital, Seoul, Korea. There were 689 patients with proven/probable invasive mould infections; of them, 46 (47 isolates) were diagnosed with NAIMIs. Fungi of the Mucorales order (n = 27, 57.4%) were the most common causative fungi, followed by Fusarium (n = 9, 19.1%). Thirty-four patients (73.9%) had neutropenia upon diagnosis of NAIMIs, and 13 (28.3%) were allogeneic stem cell transplantation recipients. The most common site of NAIMIs was the lung (n = 27, 58.7%), followed by disseminated infections (n = 8, 17.4%). There were 23.9% (n = 11) breakthrough IFIs, and 73.9% (n = 34) had co-existing bacterial or viral infections. The overall mortality at 6 and 12 weeks was 30.4% and 39.1%, respectively. Breakthrough IFIs (adjusted hazards ratio [aHR] = 1.99, 95% CI: 1.3-4.41, P = .031) and surgical treatment (aHR = 0.09, 95% CI: 0.02-0.45, P = .003) were independently associated with 6-week overall mortality. NAIMIs were not rare and occur as a complex form of infection often accompanied by breakthrough/mixed/concurrent IFIs and bacterial or viral infections. More active diagnostic efforts for NAIMIs are needed.
Collapse
Affiliation(s)
- Hyeon-Jeong Lee
- Division of Infectious Diseases, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Korea.,College of Medicine, Catholic Hematology Hospital, The Catholic University of Korea, Seoul, Korea
| | - Sung-Yeon Cho
- Division of Infectious Diseases, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Korea.,College of Medicine, Catholic Hematology Hospital, The Catholic University of Korea, Seoul, Korea.,College of Medicine, Vaccine Bio Research Institute, The Catholic University of Korea, Seoul, Korea
| | - Dong-Gun Lee
- Division of Infectious Diseases, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Korea.,College of Medicine, Catholic Hematology Hospital, The Catholic University of Korea, Seoul, Korea.,College of Medicine, Vaccine Bio Research Institute, The Catholic University of Korea, Seoul, Korea
| | - Chulmin Park
- College of Medicine, Vaccine Bio Research Institute, The Catholic University of Korea, Seoul, Korea
| | - Hye-Sun Chun
- College of Medicine, Vaccine Bio Research Institute, The Catholic University of Korea, Seoul, Korea
| | - Yeon-Joon Park
- Department of Laboratory Medicine, College of Medicine, The Catholic University of Korea, Seoul, Korea
| |
Collapse
|
46
|
Detection of Fusarium Species in Clinical Specimens by Probe-Based Real-Time PCR. J Fungi (Basel) 2019; 5:jof5040105. [PMID: 31726656 PMCID: PMC6958410 DOI: 10.3390/jof5040105] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 11/06/2019] [Accepted: 11/07/2019] [Indexed: 12/28/2022] Open
Abstract
The mold Fusarium is a ubiquitous fungus causing plant, animal and human infections. In humans, Fusarium spp. are the major cause of eye infections in patients wearing contact lenses or after local trauma. Systemic infections by Fusarium spp. mainly occur in immunosuppressed patients and can disseminate throughout the human body. Due to high levels of resistance to antifungals a fast identification of the causative agent is an urgent need. By using a probe-based real-time PCR assay specific for the genus Fusarium we analysed several different clinical specimens detecting Fusarium spp. commonly found in clinical samples in Germany. Also, a large collection of lung fluid samples of haematological patients was analysed (n = 243). In these, two samples (0.8%) were reproducibly positive, but only one could be confirmed by sequencing. For this case of probable invasive fungal disease (IFD) culture was positive for Fusarium species. Here we describe a rapid, probe-based real-time PCR assay to specifically detect DNA from a broad range of Fusarium species and its application to clinically relevant specimens.
Collapse
|
47
|
Therapeutic Challenges of Non- Aspergillus Invasive Mold Infections in Immunosuppressed Patients. Antimicrob Agents Chemother 2019; 63:AAC.01244-19. [PMID: 31481441 DOI: 10.1128/aac.01244-19] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
While Aspergillus spp. remain the major cause of invasive mold infections in hematologic cancer patients and transplant recipients, other opportunistic molds, such as Mucorales, Fusarium, and Scedosporium spp. are increasingly encountered in an expanding population of patients with severe and prolonged immunosuppression. High potential for tissue invasion and dissemination, resistance to multiple antifungals and high mortality rates are hallmarks of these non-Aspergillus invasive mold infections (NAIMIs). Assessment of drug efficacy is particularly difficult in the complex treatment scenarios of NAIMIs. Specifically, correlation between in vitro susceptibility and in vivo responses to antifungals is hard to assess, in view of the multiple, frequently interrelated factors influencing outcomes, such as pharmacokinetic/pharmacodynamic parameters determining drug availability at the site of infection, the net state of immune suppression, delay in diagnosis, or surgical debulking of infectious foci. Our current therapeutic approach of NAIMIs should evolve toward a better integration of the dynamic interactions between the pathogen, the drug and the host. Innovative concepts of experimental research may consist in manipulating the host immune system to induce a specific antifungal response or targeted drug delivery. In this review, we discuss the challenges in the management of NAIMIs and provide an update about the latest advances in diagnostic and therapeutic approaches.
Collapse
|
48
|
Closing the Gap in Surveillance and Audit of Invasive Mold Diseases for Antifungal Stewardship Using Machine Learning. J Clin Med 2019; 8:jcm8091390. [PMID: 31491944 PMCID: PMC6780614 DOI: 10.3390/jcm8091390] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2019] [Revised: 08/30/2019] [Accepted: 09/02/2019] [Indexed: 12/22/2022] Open
Abstract
Clinical audit of invasive mold disease (IMD) in hematology patients is inefficient due to the difficulties of case finding. This results in antifungal stewardship (AFS) programs preferentially reporting drug cost and consumption rather than measures that actually reflect quality of care. We used machine learning-based natural language processing (NLP) to non-selectively screen chest tomography (CT) reports for pulmonary IMD, verified by clinical review against international definitions and benchmarked against key AFS measures. NLP screened 3014 reports from 1 September 2008 to 31 December 2017, generating 784 positives that after review, identified 205 IMD episodes (44% probable-proven) in 185 patients from 50,303 admissions. Breakthrough-probable/proven-IMD on antifungal prophylaxis accounted for 60% of episodes with serum monitoring of voriconazole or posaconazole in the 2 weeks prior performed in only 53% and 69% of episodes, respectively. Fiberoptic bronchoscopy within 2 days of CT scan occurred in only 54% of episodes. The average turnaround of send-away bronchoalveolar galactomannan of 12 days (range 7–22) was associated with high empiric liposomal amphotericin consumption. A random audit of 10% negative reports revealed two clinically significant misses (0.9%, 2/223). This is the first successful use of applied machine learning for institutional IMD surveillance across an entire hematology population describing process and outcome measures relevant to AFS. Compared to current methods of clinical audit, semi-automated surveillance using NLP is more efficient and inclusive by avoiding restrictions based on any underlying hematologic condition, and has the added advantage of being potentially scalable.
Collapse
|
49
|
Cruciani M, Mengoli C, Barnes R, Donnelly JP, Loeffler J, Jones BL, Klingspor L, Maertens J, Morton CO, White LP. Polymerase chain reaction blood tests for the diagnosis of invasive aspergillosis in immunocompromised people. Cochrane Database Syst Rev 2019; 9:CD009551. [PMID: 31478559 PMCID: PMC6719256 DOI: 10.1002/14651858.cd009551.pub4] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND This is an update of the original review published in the Cochrane Database of Systematic Reviews Issue 10, 2015.Invasive aspergillosis (IA) is the most common life-threatening opportunistic invasive mould infection in immunocompromised people. Early diagnosis of IA and prompt administration of appropriate antifungal treatment are critical to the survival of people with IA. Antifungal drugs can be given as prophylaxis or empirical therapy, instigated on the basis of a diagnostic strategy (the pre-emptive approach) or for treating established disease. Consequently, there is an urgent need for research into both new diagnostic tools and drug treatment strategies. Increasingly, newer methods such as polymerase chain reaction (PCR) to detect fungal nucleic acids are being investigated. OBJECTIVES To provide an overall summary of the diagnostic accuracy of PCR-based tests on blood specimens for the diagnosis of IA in immunocompromised people. SEARCH METHODS We searched MEDLINE (1946 to June 2015) and Embase (1980 to June 2015). We also searched LILACS, DARE, Health Technology Assessment, Web of Science and Scopus to June 2015. We checked the reference lists of all the studies identified by the above methods and contacted relevant authors and researchers in the field. For this review update we updated electronic searches of the Cochrane Central Register of Controlled Trials (CENTRAL; 2018, Issue 3) in the Cochrane Library; MEDLINE via Ovid (June 2015 to March week 2 2018); and Embase via Ovid (June 2015 to 2018 week 12). SELECTION CRITERIA We included studies that: i) compared the results of blood PCR tests with the reference standard published by the European Organisation for Research and Treatment of Cancer/Mycoses Study Group (EORTC/MSG); ii) reported data on false-positive, true-positive, false-negative and true-negative results of the diagnostic tests under investigation separately; and iii) evaluated the test(s) prospectively in cohorts of people from a relevant clinical population, defined as a group of individuals at high risk for invasive aspergillosis. Case-control and retrospective studies were excluded from the analysis. DATA COLLECTION AND ANALYSIS Authors independently assessed quality and extracted data. For PCR assays, we evaluated the requirement for either one or two consecutive samples to be positive for diagnostic accuracy. We investigated heterogeneity by subgroup analyses. We plotted estimates of sensitivity and specificity from each study in receiver operating characteristics (ROC) space and constructed forest plots for visual examination of variation in test accuracy. We performed meta-analyses using the bivariate model to produce summary estimates of sensitivity and specificity. MAIN RESULTS We included 29 primary studies (18 from the original review and 11 from this update), corresponding to 34 data sets, published between 2000 and 2018 in the meta-analyses, with a mean prevalence of proven or probable IA of 16.3 (median prevalence 11.1% , range 2.5% to 57.1%). Most patients had received chemotherapy for haematological malignancy or had undergone hematopoietic stem cell transplantation. Several PCR techniques were used among the included studies. The sensitivity and specificity of PCR for the diagnosis of IA varied according to the interpretative criteria used to define a test as positive. The summary estimates of sensitivity and specificity were 79.2% (95% confidence interval (CI) 71.0 to 85.5) and 79.6% (95% CI 69.9 to 86.6) for a single positive test result, and 59.6% (95% CI 40.7 to 76.0) and 95.1% (95% CI 87.0 to 98.2) for two consecutive positive test results. AUTHORS' CONCLUSIONS PCR shows moderate diagnostic accuracy when used as screening tests for IA in high-risk patient groups. Importantly the sensitivity of the test confers a high negative predictive value (NPV) such that a negative test allows the diagnosis to be excluded. Consecutive positives show good specificity in diagnosis of IA and could be used to trigger radiological and other investigations or for pre-emptive therapy in the absence of specific radiological signs when the clinical suspicion of infection is high. When a single PCR positive test is used as the diagnostic criterion for IA in a population of 100 people with a disease prevalence of 16.3% (overall mean prevalence), three people with IA would be missed (sensitivity 79.2%, 20.8% false negatives), and 17 people would be unnecessarily treated or referred for further tests (specificity of 79.6%, 21.4% false positives). If we use the two positive test requirement in a population with the same disease prevalence, it would mean that nine IA people would be missed (sensitivity 59.6%, 40.4% false negatives) and four people would be unnecessarily treated or referred for further tests (specificity of 95.1%, 4.9% false positives). Like galactomannan, PCR has good NPV for excluding disease, but the low prevalence of disease limits the ability to rule in a diagnosis. As these biomarkers detect different markers of disease, combining them is likely to prove more useful.
Collapse
Affiliation(s)
- Mario Cruciani
- Azienda ULSS9 ScaligeraAntibiotic Stewardship ProgrammeVeronaItaly37135
| | - Carlo Mengoli
- Università di PadovaDepartment of Histology, Microbiology and Medical BiotechnologyVia Aristide Gabelli, 63PadovaItaly35121
| | - Rosemary Barnes
- Cardiff University School of MedicineInfection, Immunity and BiochemistryHeath ParkCardiffWalesUKCF14 4XN
| | - J Peter Donnelly
- Nijmegen Institute for InfectionDepartment of HaematologyInflammation and ImmunityRadboud University Nijmegen Medical CenterNijmegenNetherlands
| | - Juergen Loeffler
- Julius‐Maximilians‐UniversitatMedizinische Klinik IIKlinikstrasse 6‐8WurzburgGermany97070
| | - Brian L Jones
- Glasgow Royal Infirmary & University of GlasgowDepartment of Medical MicrobiologyGlasgowUK
| | - Lena Klingspor
- Division of Clinical MicrobiologyDepartment of Laboratory MedicineKarolinska University HospitalStockholmSweden
| | - Johan Maertens
- Acute Leukemia and Stem Cell Transplantation UnitDepartment of HematologyUniversity Hospitals LeuvenLeuvenBelgium
| | - Charles O Morton
- Western Sydney UniversitySchool of Science and HealthCampbelltown CampusCampbelltownNew South WalesAustralia2560
| | - Lewis P White
- Microbiology Cardiff, UHWPublic Health WalesHeath ParkCardiffUKCF37 1EN
| | | |
Collapse
|
50
|
Cornely OA, Hoenigl M, Lass-Flörl C, Chen SCA, Kontoyiannis DP, Morrissey CO, Thompson GR. Defining breakthrough invasive fungal infection-Position paper of the mycoses study group education and research consortium and the European Confederation of Medical Mycology. Mycoses 2019; 62:716-729. [PMID: 31254420 PMCID: PMC6692208 DOI: 10.1111/myc.12960] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 06/26/2019] [Accepted: 06/27/2019] [Indexed: 12/14/2022]
Abstract
Breakthrough invasive fungal infections (IFIs) have emerged as a significant problem in patients receiving systemic antifungals; however, consensus criteria for defining breakthrough IFI are missing. This position paper establishes broadly applicable definitions of breakthrough IFI for clinical research. Representatives of the Mycoses Study Group Education and Research Consortium (MSG-ERC) and the European Confederation of Medical Mycology (ECMM) reviewed the relevant English literature for definitions applied and published through 2018. A draft proposal for definitions was developed and circulated to all members of the two organisations for comment and suggestions. The authors addressed comments received and circulated the updated document for approval. Breakthrough IFI was defined as any IFI occurring during exposure to an antifungal drug, including fungi outside the spectrum of activity of an antifungal. The time of breakthrough IFI was defined as the first attributable clinical sign or symptom, mycological finding or radiological feature. The period defining breakthrough IFI depends on pharmacokinetic properties and extends at least until one dosing interval after drug discontinuation. Persistent IFI describes IFI that is unchanged/stable since treatment initiation with ongoing need for antifungal therapy. It is distinct from refractory IFI, defined as progression of disease and therefore similar to non-response to treatment. Relapsed IFI occurs after treatment and is caused by the same pathogen at the same site, although dissemination can occur. These proposed definitions are intended to support the design of future clinical trials and epidemiological research in clinical mycology, with the ultimate goal of increasing the comparability of clinical trial results.
Collapse
Affiliation(s)
- Oliver A Cornely
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
- Department I of Internal Medicine, ECMM Center of Excellence for Medical Mycology, German Centre for Infection Research, Partner Site Bonn-Cologne (DZIF), University of Cologne, Cologne, Germany
- Clinical Trials Centre Cologne (ZKS Köln), University of Cologne, Cologne, Germany
| | - Martin Hoenigl
- Division of Infectious Diseases, University of California San Diego, San Diego, CA, USA
- Division of Pulmonology and Section of Infectious Diseases, Medical University of Graz, Graz, Austria
| | - Cornelia Lass-Flörl
- Division of Hygiene and Microbiology, ECMM Excellence Center for Medical Mycology, Medical University Innsbruck, Innsbruck, Austria
| | - Sharon C-A Chen
- Centre for Infectious Diseases and Microbiology, Laboratory Services, ICPMR, New South Wales Health Pathology, Westmead Hospital, Centre for Infectious Diseases and Microbiology, Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
| | - Dimitrios P Kontoyiannis
- Department of Infectious Diseases, Infection Control, and Employee Health, University of Texas M. D. Anderson Cancer Center, Houston, TX, USA
| | - C Orla Morrissey
- Infectious Diseases, Alfred Health and Monash University, Melbourne, VIC, Australia
| | - George R Thompson
- Departments of Medical Microbiology and Immunology and Internal Medicine Division of Infectious Diseases, UC-Davis Medical Center, Sacramento, CA, USA
| |
Collapse
|