1
|
Regan J, DeJarnette C, Reitler P, Gihaz S, Srivastava A, Ge W, Tucker KM, Peters TL, Meibohm B, Ben Mamoun C, Fortwendel JR, Hevener KE, Palmer GE. Pantothenate kinase is an effective target for antifungal therapy. Cell Chem Biol 2025; 32:710-721.e6. [PMID: 40378822 DOI: 10.1016/j.chembiol.2025.04.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 02/11/2025] [Accepted: 04/14/2025] [Indexed: 05/19/2025]
Abstract
Pantothenate kinase (PanK) catalyzes the first step in the conversion of pantothenate to coenzyme A (CoA), an essential cofactor in all living organisms. The findings of this study demonstrate that PanK is essential for the viability and virulence of two of the most medically significant fungi-the pathogenic yeast Candida albicans, and the infectious mold Aspergillus fumigatus-within the mammalian host. Biochemical, biophysical as well as chemical-genetic approaches were applied to identify 3,4-methylenedioxy-β-nitrostyrene (MNS) as a broad-spectrum antifungal that directly engages and inhibits PanK to block CoA production. Importantly, MNS is inactive against a mammalian PanK and demonstrates in vivo antifungal efficacy a mouse model of disseminated C. albicans infection. Thus, MNS has provided a valuable chemical probe to establish the validity of targeting PanK with small molecule inhibitors as a strategy to develop efficacious antifungal therapeutics.
Collapse
Affiliation(s)
- Jessica Regan
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Sciences Center, Memphis, TN, USA
| | - Christian DeJarnette
- Department of Clinical Pharmacy and Translational Science, College of Pharmacy, University of Tennessee Health Sciences Center, Memphis, TN, USA
| | - Parker Reitler
- Department of Molecular Immunology and Biochemistry, College of Graduate Health Sciences, University of Tennessee Health Sciences Center, Memphis, TN, USA
| | - Shalev Gihaz
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Ashish Srivastava
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Sciences Center, Memphis, TN, USA
| | - Wenbo Ge
- Department of Clinical Pharmacy and Translational Science, College of Pharmacy, University of Tennessee Health Sciences Center, Memphis, TN, USA
| | - Katie M Tucker
- Department of Clinical Pharmacy and Translational Science, College of Pharmacy, University of Tennessee Health Sciences Center, Memphis, TN, USA
| | - Tracy L Peters
- Department of Clinical Pharmacy and Translational Science, College of Pharmacy, University of Tennessee Health Sciences Center, Memphis, TN, USA
| | - Bernd Meibohm
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Sciences Center, Memphis, TN, USA
| | - Choukri Ben Mamoun
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Jarrod R Fortwendel
- Department of Clinical Pharmacy and Translational Science, College of Pharmacy, University of Tennessee Health Sciences Center, Memphis, TN, USA
| | - Kirk E Hevener
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Sciences Center, Memphis, TN, USA.
| | - Glen E Palmer
- Department of Clinical Pharmacy and Translational Science, College of Pharmacy, University of Tennessee Health Sciences Center, Memphis, TN, USA.
| |
Collapse
|
2
|
Reitler P, DeJarnette CA, Kumar R, Tucker KM, Peters TL, Twarog NR, Shelat AA, Palmer GE. A screen to identify antifungal antagonists reveals a variety of pharmacotherapies induce echinocandin tolerance in Candida albicans. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.18.638903. [PMID: 40027746 PMCID: PMC11870487 DOI: 10.1101/2025.02.18.638903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Through screening a comprehensive collection of drugs approved for human use, we identified over 20 that oppose the antifungal activity of the echinocandins upon the infectious yeast, Candida albicans . More detailed evaluation of five such drugs, including the atypical antipsychotic aripiprazole and the tyrosine kinase inhibitor ponatinib, indicated they promote C. albicans survival following exposure to the echinocandin antifungals. The activity of the five selected antagonists was dependent upon the Mkc1p MAPK pathway, however, ponatinib was paradoxically shown to suppress phosphorylation and therefore activation of Mkc1p itself. Components of several other signaling pathways are also required, including those of calcineurin and casein kinase-2, suggesting the observed antagonism required much of the cell wall stress responses previously described for C. albicans . Transcriptome analysis revealed that the antagonists stimulated the expression of genes involved in xenobiotic and antifungal resistance, and suppressed the expression of genes associated with hyphal growth. Thus, the echinocandin antagonistic drugs modulate C. albicans physiology in ways that could impact its pathogenicity and/or response to therapeutic intervention. Finally, a mutant lacking the Efg1p transcription factor, which has a central role in the activation of C. albicans hyphal growth was found to have intrinsically high levels of echinocandin tolerance, suggesting a link between modulation of morphogenesis related signaling and echinocandin tolerance. Importance We report a substantial number of previously unknown drug interactions that modulate the echinocandin sensitivity of one of the most prevalent human fungal pathogens, Candida albicans . The echinocandins are the first line therapy for treating disseminated and often lethal Candida infections, that account for >75% of invasive fungal infections in the U.S.. For largely unknown reasons, a substantial number of patients with invasive candidiasis fail to respond to treatment with these drugs. The finding of this study suggest that co-administered medications have the potential to influence the therapeutic outcomes of invasive fungal infections through modulating antifungal drug tolerance and/or fungal pathogenicity. The potential for echinocandin antagonistic medications to influence therapeutic outcomes is discussed.
Collapse
Affiliation(s)
- Parker Reitler
- Department of Clinical Pharmacy and Translational Science, College of Pharmacy, University of Tennessee Health Sciences Center, Memphis, USA
| | - Christian A. DeJarnette
- Department of Clinical Pharmacy and Translational Science, College of Pharmacy, University of Tennessee Health Sciences Center, Memphis, USA
| | - Ravinder Kumar
- Department of Clinical Pharmacy and Translational Science, College of Pharmacy, University of Tennessee Health Sciences Center, Memphis, USA
| | - Katie M. Tucker
- Department of Clinical Pharmacy and Translational Science, College of Pharmacy, University of Tennessee Health Sciences Center, Memphis, USA
| | - Tracy L. Peters
- Department of Pharmacy and Pharmaceutical Sciences, St Jude Children’s Research Hospital, Memphis, TN, USA
| | - Nathaniel R Twarog
- Department of Chemical Biology and Therapeutics, St Jude Children’s Research Hospital, Memphis, TN, USA
| | - Anang A. Shelat
- Department of Chemical Biology and Therapeutics, St Jude Children’s Research Hospital, Memphis, TN, USA
| | - Glen E. Palmer
- Department of Clinical Pharmacy and Translational Science, College of Pharmacy, University of Tennessee Health Sciences Center, Memphis, USA
| |
Collapse
|
3
|
Zhai B, Liao C, Jaggavarapu S, Tang Y, Rolling T, Ning Y, Sun T, Bergin SA, Gjonbalaj M, Miranda E, Babady NE, Bader O, Taur Y, Butler G, Zhang L, Xavier JB, Weiss DS, Hohl TM. Antifungal heteroresistance causes prophylaxis failure and facilitates breakthrough Candida parapsilosis infections. Nat Med 2024; 30:3163-3172. [PMID: 39095599 PMCID: PMC11840754 DOI: 10.1038/s41591-024-03183-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Accepted: 07/08/2024] [Indexed: 08/04/2024]
Abstract
Breakthrough fungal infections in patients on antimicrobial prophylaxis during allogeneic hematopoietic cell transplantation (allo-HCT) represent a major and often unexplained cause of morbidity and mortality. Candida parapsilosis is a common cause of invasive candidiasis and has been classified as a high-priority fungal pathogen by the World Health Organization. In high-risk allo-HCT recipients on micafungin prophylaxis, we show that heteroresistance (the presence of a phenotypically unstable, low-frequency subpopulation of resistant cells (~1 in 10,000)) underlies breakthrough bloodstream infections by C. parapsilosis. By analyzing 219 clinical isolates from North America, Europe and Asia, we demonstrate widespread micafungin heteroresistance in C. parapsilosis. Standard antimicrobial susceptibility tests, such as broth microdilution or gradient diffusion assays, which guide drug selection for invasive infections, fail to detect micafungin heteroresistance in C. parapsilosis. To facilitate rapid detection of micafungin heteroresistance in C. parapsilosis, we constructed a predictive machine learning framework that classifies isolates as heteroresistant or susceptible using a maximum of ten genomic features. These results connect heteroresistance to unexplained antifungal prophylaxis failure in allo-HCT recipients and demonstrate a proof-of-principle diagnostic approach with the potential to guide clinical decisions and improve patient care.
Collapse
Affiliation(s)
- Bing Zhai
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.
- Infectious Disease Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| | - Chen Liao
- Computational and Systems Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Siddharth Jaggavarapu
- Emory Antibiotic Resistance Center, Atlanta, GA, USA
- Department of Microbiology and Immunology, Emory University, Atlanta, GA, USA
- Emory Vaccine Center, Atlanta, GA, USA
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Yuanyuan Tang
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Thierry Rolling
- Infectious Disease Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Yating Ning
- Department of Laboratory Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
- Beijing Key Laboratory for Mechanisms Research and Precision Diagnosis of Invasive Fungal Diseases, Beijing, China
| | - Tianshu Sun
- Beijing Key Laboratory for Mechanisms Research and Precision Diagnosis of Invasive Fungal Diseases, Beijing, China
- Clinical Biobank, Medical Research Center, National Science and Technology Key Infrastructure on Translational Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Sean A Bergin
- School of Biomolecular and Biomedical Science, Conway Institute, University College Dublin, Belfield, Dublin, Ireland
| | - Mergim Gjonbalaj
- Infectious Disease Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Edwin Miranda
- Clinical Microbiology Service, Department of Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - N Esther Babady
- Infectious Disease Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Clinical Microbiology Service, Department of Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Oliver Bader
- Institute for Medical Microbiology and Virology, University Medical Center Göttingen, Göttingen, Germany
| | - Ying Taur
- Infectious Disease Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Geraldine Butler
- School of Biomolecular and Biomedical Science, Conway Institute, University College Dublin, Belfield, Dublin, Ireland
| | - Li Zhang
- Department of Laboratory Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
- Beijing Key Laboratory for Mechanisms Research and Precision Diagnosis of Invasive Fungal Diseases, Beijing, China
| | - Joao B Xavier
- Computational and Systems Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - David S Weiss
- Emory Antibiotic Resistance Center, Atlanta, GA, USA.
- Department of Microbiology and Immunology, Emory University, Atlanta, GA, USA.
- Emory Vaccine Center, Atlanta, GA, USA.
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA.
| | - Tobias M Hohl
- Infectious Disease Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA.
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
4
|
Taynton T, Allsup D, Barlow G. How can we optimize antifungal use and stewardship in the treatment of acute leukemia? Expert Rev Hematol 2024; 17:581-593. [PMID: 39037307 DOI: 10.1080/17474086.2024.2383401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 07/18/2024] [Indexed: 07/23/2024]
Abstract
INTRODUCTION The global need for antifungal stewardship is driven by spreading antimicrobial and antifungal resistance. Triazoles are the only oral and relatively well-tolerated class of antifungal medications, and usage is associated with acquired resistance and species replacement with intrinsically resistant organisms. On a per-patient basis, hematology patients are the largest inpatient consumers of antifungal drugs, but are also the most vulnerable to invasive fungal disease. AREAS COVERED In this review we discuss available and forthcoming antifungal drugs, antifungal prophylaxis and empiric antifungal therapy, and how a screening based and diagnostic-driven approach may be used to reduce antifungal consumption. Finally, we discuss components of an antifungal stewardship program, interventions that can be employed, and how impact can be measured. The search methodology consisted of searching PubMed for journal articles using the term antifungal stewardship plus program, intervention, performance measure or outcome before 1 January 2024. EXPERT OPINION Initial focus should be on implementing effective antifungal stewardship programs by developing and implementing local guidelines and using interventions, such as post-prescription review and feedback, which are known to be effective. Technologies such as microbiome analysis and machine learning may allow the development of truly individualized risk-factor-based approaches to antifungal stewardship in the future.
Collapse
Affiliation(s)
- Thomas Taynton
- Department of Infection, Hull University Teaching Hospitals NHS Trust, Hull, UK
- Centre for Biomedical Research, Hull York Medical School, Hull, UK
| | - David Allsup
- Biomedical Institute for Multimorbidity, Hull York Medical School, Hull, UK
- Queen's Centre for Oncology and Haematology, Hull University Teaching Hospitals NHS Trust, Hull, UK
| | - Gavin Barlow
- Department of Infection, Hull University Teaching Hospitals NHS Trust, Hull, UK
- York Biomedical Research Institute and Hull York Medical School, University of York, York, UK
| |
Collapse
|
5
|
Panagopoulou P, Roilides E. An update on pharmacotherapy for fungal infections in allogeneic stem cell transplant recipients. Expert Opin Pharmacother 2024; 25:1453-1482. [PMID: 39096057 DOI: 10.1080/14656566.2024.2387686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Revised: 07/24/2024] [Accepted: 07/30/2024] [Indexed: 08/04/2024]
Abstract
INTRODUCTION Invasive fungal diseases (IFD) constitute a major cause of morbidity and mortality in hematopoietic stem cell transplantation (HSCT) recipients. AREAS COVERED We describe epidemiology, causes and risk factors of IFD in allogeneic HSCT discussing prophylaxis and treatment in various HSCT phases. We present the most recent studies on this thematic area, including novel data on currently available antifungals, i.e. formulations, dosing, safety, efficacy and therapeutic drug monitoring. Finally, we present the most recent relevant recommendations published. Literature search included PubMed, Scopus, and clinicaltrials.gov between January 2014 and April 2024. EXPERT OPINION The antifungal agents employed for prophylaxis and therapy should be predicated on local epidemiology of IFD. Fluconazole prophylaxis remains a first-line choice before engraftment when the main pathogen is Candida spp. After engraftment, prophylaxis should be with mold-active agents (i.e. triazoles). For candidiasis, echinocandins are suggested as first-line treatment, whereas aspergillosis responds well to mold-active azoles and liposomal amphotericin B (L-AmB). For mucormycosis, treatment of choice includes L-AmB and isavuconazole. Choice between fever-driven and diagnostics-driven strategies remains equivocal. Open research topics remain: 1) optimization of tools to ensure prompt and accurate IFD diagnosis to avoid unnecessary exposure to antifungals, drug interactions and cost; 2) refinement of treatment for resistant/refractory strains.
Collapse
Affiliation(s)
- Paraskevi Panagopoulou
- Pediatric Hematology & Oncology, 4th Department of Pediatrics, Aristotle University School of Medicine and Papageorgiou General Hospital, Thessaloniki, Greece
| | - Emmanuel Roilides
- Infectious Diseases Unit, 3rd Department of Pediatrics, Aristotle University School of Medicine and Hippokration General Hospital, Thessaloniki, Greece
| |
Collapse
|
6
|
Khilnani GC, Tiwari P, Mittal S, Kulkarni AP, Chaudhry D, Zirpe KG, Todi SK, Mohan A, Hegde A, Jagiasi BG, Krishna B, Rodrigues C, Govil D, Pal D, Divatia JV, Sengar M, Gupta M, Desai M, Rungta N, Prayag PS, Bhattacharya PK, Samavedam S, Dixit SB, Sharma S, Bandopadhyay S, Kola VR, Deswal V, Mehta Y, Singh YP, Myatra SN. Guidelines for Antibiotics Prescription in Critically Ill Patients. Indian J Crit Care Med 2024; 28:S104-S216. [PMID: 39234229 PMCID: PMC11369928 DOI: 10.5005/jp-journals-10071-24677] [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: 02/14/2024] [Accepted: 03/20/2024] [Indexed: 09/06/2024] Open
Abstract
How to cite this article: Khilnani GC, Tiwari P, Mittal S, Kulkarni AP, Chaudhry D, Zirpe KG, et al. Guidelines for Antibiotics Prescription in Critically Ill Patients. Indian J Crit Care Med 2024;28(S2):S104-S216.
Collapse
Affiliation(s)
- Gopi C Khilnani
- Department of Pulmonary, Critical Care and Sleep Medicine, PSRI Hospital, New Delhi, India
| | - Pawan Tiwari
- Department of Pulmonary, Critical Care and Sleep Medicine, AIIMS, New Delhi, India
| | - Saurabh Mittal
- Department of Pulmonary, Critical Care and Sleep Medicine, AIIMS, New Delhi, India
| | - Atul P Kulkarni
- Division of Critical Care Medicine, Department of Anaesthesia, Critical Care and Pain, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Dhruva Chaudhry
- Department of Pulmonary and Critical Care Medicine, University of Health Sciences, Rohtak, Haryana, India
| | - Kapil G Zirpe
- Department of Neuro Trauma Unit, Grant Medical Foundation, Pune, Maharashtra, India
| | - Subhash K Todi
- Department of Critical Care, AMRI Hospital, Kolkata, West Bengal, India
| | - Anant Mohan
- Department of Pulmonary, Critical Care and Sleep Medicine, AIIMS, New Delhi, India
| | - Ashit Hegde
- Department of Medicine & Critical Care, P D Hinduja National Hospital, Mumbai, India
| | - Bharat G Jagiasi
- Department of Critical Care, Kokilaben Dhirubhai Ambani Hospital, Navi Mumbai, Maharashtra, India
| | - Bhuvana Krishna
- Department of Critical Care Medicine, St John's Medical College and Hospital, Bengaluru, India
| | - Camila Rodrigues
- Department of Microbiology, P D Hinduja National Hospital, Mumbai, India
| | - Deepak Govil
- Department of Critical Care and Anesthesia, Medanta – The Medicity, GuruGram, Haryana, India
| | - Divya Pal
- Department of Critical Care and Anesthesia, Medanta – The Medicity, GuruGram, Haryana, India
| | - Jigeeshu V Divatia
- Department of Anaesthesiology, Critical Care and Pain, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Manju Sengar
- Department of Medical Oncology, Tata Memorial Center, Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Mansi Gupta
- Department of Pulmonary Medicine, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, Uttar Pradesh, India
| | - Mukesh Desai
- Department of Immunology, Pediatric Hematology and Oncology Bai Jerbai Wadia Hospital for Children, Mumbai, Maharashtra, India
| | - Narendra Rungta
- Department of Critical Care & Anaesthesiology, Rajasthan Hospital, Jaipur, India
| | - Parikshit S Prayag
- Department of Transplant Infectious Diseases, Deenanath Mangeshkar Hospital, Pune, Maharashtra, India
| | - Pradip K Bhattacharya
- Department of Critical Care Medicine, Rajendra Institute of Medical Sciences, Ranchi, Jharkhand, India
| | - Srinivas Samavedam
- Department of Critical Care, Ramdev Rao Hospital, Hyderabad, Telangana, India
| | - Subhal B Dixit
- Department of Critical Care, Sanjeevan and MJM Hospital, Pune, Maharashtra, India
| | - Sudivya Sharma
- Department of Anaesthesiology, Critical Care and Pain, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Susruta Bandopadhyay
- Department of Critical Care, AMRI Hospitals Salt Lake, Kolkata, West Bengal, India
| | - Venkat R Kola
- Department of Critical Care Medicine, Yashoda Hospitals, Hyderabad, Telangana, India
| | - Vikas Deswal
- Consultant, Infectious Diseases, Medanta - The Medicity, Gurugram, Haryana, India
| | - Yatin Mehta
- Department of Critical Care and Anesthesia, Medanta – The Medicity, GuruGram, Haryana, India
| | - Yogendra P Singh
- Department of Critical Care, Max Super Speciality Hospital, Patparganj, New Delhi, India
| | - Sheila N Myatra
- Department of Anaesthesiology, Critical Care and Pain, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, Maharashtra, India
| |
Collapse
|
7
|
Ueda T, Nakajima K, Ichiki K, Ishikawa K, Yamada K, Tsuchida T, Otani N, Takubo S, Iijima K, Uchino M, Horio Y, Kuwahara R, Kimura T, Murakami Y, Nozaki Y, Nakama S, Miyazaki Y, Takesue Y. Association between the hypokalaemia index based on area over the serum potassium concentration curve and occurrence of acute kidney injury in patients administered liposomal amphotericin B. Mycoses 2024; 67:e13771. [PMID: 39031945 DOI: 10.1111/myc.13771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 07/11/2024] [Accepted: 07/13/2024] [Indexed: 07/22/2024]
Abstract
BACKGROUND Acute kidney injury (AKI) and hypokalaemia are common adverse events after treatment with liposomal amphotericin B (L-AMB). OBJECTIVES Because excess potassium (K) leakage occurs during renal tubular injury caused by L-AMB, measuring the decrease in rate of serum K concentration might be more useful to assess the renal impact of L-AMB than hypokalaemia identified from a one-point measurement. The effects of a decrease in K concentration and duration of hypokalaemia on AKI were investigated. METHODS A ≥ 10% decrease in K concentration from the reference concentration within a 7-day timeframe was evaluated. The hypokalaemia index, which combines the duration of K concentration lower than the reference and a marked low K concentration, was calculated from the area over the concentration curve. RESULTS Eighty-six patients were included in the study. The incidences of AKI and decrease in K concentration were 36.0% and 63.9%, respectively. Of patients who developed both adverse events, a decrease in K concentration occurred first in 22 of 26 patients, followed by AKI 7 days later. Hypokalaemia did not increase AKI risk whereas a decrease in K concentration was an independent risk factor for AKI. The hypokalaemia index in patients with AKI was significantly higher than those without AKI (5.35 vs. 2.50 points, p = 0.002), and ≥3.45 points was a significant predictor for AKI. CONCLUSION A ≥ 10% decrease in the K concentration was a significant factor for AKI in patients receiving L-AMB therapy. In such patients, dose reduction or alternative antifungals could be considered based on the hypokalaemia index.
Collapse
Affiliation(s)
- Takashi Ueda
- Department of Infection Prevention and Control, Hyogo Medical University Hospital, Nishinomiya, Japan
| | - Kazuhiko Nakajima
- Department of Infection Prevention and Control, Hyogo Medical University Hospital, Nishinomiya, Japan
| | - Kaoru Ichiki
- Department of Infection Prevention and Control, Hyogo Medical University Hospital, Nishinomiya, Japan
| | - Kaori Ishikawa
- Department of Infection Prevention and Control, Hyogo Medical University Hospital, Nishinomiya, Japan
| | - Kumiko Yamada
- Department of Infection Prevention and Control, Hyogo Medical University Hospital, Nishinomiya, Japan
- Department of Clinical Technology, Hyogo Medical University Hospital, Nishinomiya, Japan
| | - Toshie Tsuchida
- Department of Infection Prevention and Control, Hyogo Medical University Hospital, Nishinomiya, Japan
| | - Naruhito Otani
- Department of Infection Prevention and Control, Hyogo Medical University Hospital, Nishinomiya, Japan
| | - Shingo Takubo
- Department of Pharmacy, Hyogo Medical University Hospital, Nishinomiya, Japan
| | - Kosuke Iijima
- Department of Clinical Technology, Hyogo Medical University Hospital, Nishinomiya, Japan
| | - Motoi Uchino
- Division of Inflammatory Bowel Disease Surgery, Department of Gastroenterological Surgery, Hyogo Medical University, Nishinomiya, Japan
| | - Yuki Horio
- Division of Inflammatory Bowel Disease Surgery, Department of Gastroenterological Surgery, Hyogo Medical University, Nishinomiya, Japan
| | - Ryuichi Kuwahara
- Division of Inflammatory Bowel Disease Surgery, Department of Gastroenterological Surgery, Hyogo Medical University, Nishinomiya, Japan
| | - Takeshi Kimura
- Department of Pharmacy, Hyogo Medical University Hospital, Nishinomiya, Japan
| | - Yasushi Murakami
- Department of Respiratory Medicine, Tokoname City Hospital, Tokoname, Japan
| | - Yasuhiro Nozaki
- Department of Respiratory Medicine, Tokoname City Hospital, Tokoname, Japan
| | - Soichiro Nakama
- Department of Clinical Infectious Diseases, Tokoname City Hospital, Tokoname, Japan
| | - Yoshitsugu Miyazaki
- Department of Chemotherapy and Mycoses, National Institute of Infectious Diseases, Tokyo, Japan
| | - Yoshio Takesue
- Department of Infection Prevention and Control, Hyogo Medical University Hospital, Nishinomiya, Japan
- Department of Clinical Infectious Diseases, Tokoname City Hospital, Tokoname, Japan
| |
Collapse
|
8
|
Akinosoglou K, Rigopoulos EA, Papageorgiou D, Schinas G, Polyzou E, Dimopoulou E, Gogos C, Dimopoulos G. Amphotericin B in the Era of New Antifungals: Where Will It Stand? J Fungi (Basel) 2024; 10:278. [PMID: 38667949 PMCID: PMC11051097 DOI: 10.3390/jof10040278] [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: 03/19/2024] [Revised: 04/05/2024] [Accepted: 04/07/2024] [Indexed: 04/28/2024] Open
Abstract
Amphotericin B (AmB) has long stood as a cornerstone in the treatment of invasive fungal infections (IFIs), especially among immunocompromised patients. However, the landscape of antifungal therapy is evolving. New antifungal agents, boasting novel mechanisms of action and better safety profiles, are entering the scene, presenting alternatives to AmB's traditional dominance. This shift, prompted by an increase in the incidence of IFIs, the growing demographic of immunocompromised individuals, and changing patterns of fungal resistance, underscores the continuous need for effective treatments. Despite these challenges, AmB's broad efficacy and low resistance rates maintain its essential status in antifungal therapy. Innovations in AmB formulations, such as lipid complexes and liposomal delivery systems, have significantly mitigated its notorious nephrotoxicity and infusion-related reactions, thereby enhancing its clinical utility. Moreover, AmB's efficacy in treating severe and rare fungal infections and its pivotal role as prophylaxis in high-risk settings highlight its value and ongoing relevance. This review examines AmB's standing amidst the ever-changing antifungal landscape, focusing on its enduring significance in current clinical practice and exploring its potential future therapeutic adaptations.
Collapse
Affiliation(s)
- Karolina Akinosoglou
- School of Medicine, University of Patras, 26504 Patras, Greece; (E.A.R.); (D.P.); (G.S.); (E.P.); (C.G.)
- Department of Internal Medicine and Infectious Diseases, University General Hospital of Patras, 26504 Rio, Greece
| | | | - Despoina Papageorgiou
- School of Medicine, University of Patras, 26504 Patras, Greece; (E.A.R.); (D.P.); (G.S.); (E.P.); (C.G.)
| | - Georgios Schinas
- School of Medicine, University of Patras, 26504 Patras, Greece; (E.A.R.); (D.P.); (G.S.); (E.P.); (C.G.)
| | - Eleni Polyzou
- School of Medicine, University of Patras, 26504 Patras, Greece; (E.A.R.); (D.P.); (G.S.); (E.P.); (C.G.)
| | | | - Charalambos Gogos
- School of Medicine, University of Patras, 26504 Patras, Greece; (E.A.R.); (D.P.); (G.S.); (E.P.); (C.G.)
| | - George Dimopoulos
- 3rd Department of Critical Care, Evgenidio Hospital, Medical School, National and Kapodistrian University of Athens, 11528 Athens, Greece;
| |
Collapse
|
9
|
Araujo GLV, Murta Amaral L, Ponzio V, Rocha JL. Economic and budgetary impact evaluation of isavuconazole (Cresemba®) versus voriconazole (Vfend®) for the treatment of patients with possible invasive aspergillosis from the perspective of the Brazilian supplementary health system. PLoS One 2024; 19:e0299056. [PMID: 38427635 PMCID: PMC10906891 DOI: 10.1371/journal.pone.0299056] [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: 08/28/2023] [Accepted: 02/04/2024] [Indexed: 03/03/2024] Open
Abstract
OBJECTIVES This study aims to evaluate the cost-utility and the budgetary impact of isavuconazole compared to voriconazole in patients with suspected invasive aspergillosis (IA) from the perspective of the Brazilian supplementary health system (SHS). METHODS In this model, a decision tree was developed and included patients with possible IA. Efficacy parameters were extracted from the clinical studies. Drug acquisition, hospitalization costs and adverse events were also collected. Alternative 3- and 10-year time horizon scenarios were used. In addition, deterministic and probabilistic sensitivity analyses were simulated. A budget impact analysis of isavuconazole versus voriconazole was performed, assuming a time horizon of 5 years. In addition, sensitivity analyses were conducted to assess the robustness of the model. Results are reported in Brazilian Real (BRL), year values 2022. RESULTS The economic analysis of the base case showed that isavuconazole is associated with a saving of 95,174.00 BRL per patient compared to voriconazole. All other simulated scenarios showed that isavuconazole is dominant versus comparators when considering a willingness to pay 40,688.00 BRL/Quality-Adjusted Life Years (QALY). The results were considered robust by the sensitivity analyses. The budget impact analysis showed that the incorporation of isavuconazole generates savings to the SHS, compared to voriconazole, of approximately 20.5 million BRL in the first year. This reaches about 54 million BRL in the fifth incorporation year, considering the market penetration of 20% in the first year, and 50% in the fifth year. CONCLUSION Compared with voriconazole, isavuconazole is regarded as a dominant treatment strategy for patients with suspected IA and generates savings for the SHS.
Collapse
Affiliation(s)
| | | | | | - Jaime Luis Rocha
- Faculty of Medicine of the Pontifical Catholic University of Paraná, PUC, Paraná, Brazil
| |
Collapse
|
10
|
Andreev SS, Bronin GO, Epifanova NY, Kozlova OP, Pristanskova EA, Khostelidi SN, Shadrivova OV. Benefits of early antifungal therapy in hematology patients. ONCOHEMATOLOGY 2024; 19:99-112. [DOI: 10.17650/1818-8346-2024-19-1-99-112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
Invasive fungal infections (IFIs) are a serious threat to patients with hematological diseases. These infections are characterized by high mortality and lead to significant financial costs for treatment. The most common pathogens of IFIs are Aspergillus spp. and Candida spp., but in recent years, cases of infections caused by rare pathogens have become more frequent. Diagnosis of IFIs and choice of treatment remain challenging due to the nonspecificity of symptoms and the diversity of clinical cases. In this regard, the problem of start time and choice of antifungal therapy remains of current interest. This review briefly describes diagnostic criteria, challenges associated with IFIs diagnosing, provides evidence for empiric and preventive strategies as two early treatment approaches, and examines the impact of therapy initiation on patient outcomes. Treatment of IFIs in hematologic patients should be individualized. At the same time, early administration of therapy with broad-spectrum drugs for febrile neutropenia and parallel diagnostic measures can improve treatment outcomes. There is a lack of current data on the benefits of specific treatment strategies, highlighting the need for further research.
Collapse
Affiliation(s)
| | - G. O. Bronin
- Morozov Children’s City Clinical Hospital of the Moscow Healthcare Department
| | - N. Yu. Epifanova
- N.N. Blokhin National Medical Research Center of Oncology, Ministry of Health of Russia
| | - O. P. Kozlova
- North-Western State Medical University named after I.I. Mechnikov, Ministry of Health of Russia
| | - E. A. Pristanskova
- Russian Children’s Clinical Hospital ‒ branch of the N.I. Pirogov Russian National Research Medical University, Ministry of Health of Russia
| | - S. N. Khostelidi
- North-Western State Medical University named after I.I. Mechnikov, Ministry of Health of Russia
| | - O. V. Shadrivova
- North-Western State Medical University named after I.I. Mechnikov, Ministry of Health of Russia
| |
Collapse
|
11
|
Sprute R, Nacov JA, Neofytos D, Oliverio M, Prattes J, Reinhold I, Cornely OA, Stemler J. Antifungal prophylaxis and pre-emptive therapy: When and how? Mol Aspects Med 2023; 92:101190. [PMID: 37207579 DOI: 10.1016/j.mam.2023.101190] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Revised: 04/22/2023] [Accepted: 05/05/2023] [Indexed: 05/21/2023]
Abstract
The growing pool of critically ill or immunocompromised patients leads to a constant increase of life-threatening invasive infections by fungi such as Aspergillus spp., Candida spp. and Pneumocystis jirovecii. In response to this, prophylactic and pre-emptive antifungal treatment strategies have been developed and implemented for high-risk patient populations. The benefit by risk reduction needs to be carefully weighed against potential harm caused by prolonged exposure against antifungal agents. This includes adverse effects and development of resistance as well as costs for the healthcare system. In this review, we summarise evidence and discuss advantages and downsides of antifungal prophylaxis and pre-emptive treatment in the setting of malignancies such as acute leukaemia, haematopoietic stem cell transplantation, CAR-T cell therapy, and solid organ transplant. We also address preventive strategies in patients after abdominal surgery and with viral pneumonia as well as individuals with inherited immunodeficiencies. Notable progress has been made in haematology research, where strong recommendations regarding antifungal prophylaxis and pre-emptive treatment are backed by data from randomized controlled trials, whereas other critical areas still lack high-quality evidence. In these areas, paucity of definitive data translates into centre-specific strategies that are based on interpretation of available data, local expertise, and epidemiology. The development of novel immunomodulating anticancer drugs, high-end intensive care treatment and the development of new antifungals with new modes of action, adverse effects and routes of administration will have implications on future prophylactic and pre-emptive approaches.
Collapse
Affiliation(s)
- Rosanne Sprute
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Translational Research, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne, Germany; University of Cologne, Faculty of Medicine and University Hospital Cologne, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD) and Excellence Center for Medical Mycology (ECMM), Cologne, Germany; German Centre for Infection Research (DZIF), Partner Site Bonn-Cologne, Cologne, Germany
| | - Julia A Nacov
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Translational Research, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne, Germany; University of Cologne, Faculty of Medicine and University Hospital Cologne, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD) and Excellence Center for Medical Mycology (ECMM), Cologne, Germany; German Centre for Infection Research (DZIF), Partner Site Bonn-Cologne, Cologne, Germany
| | - Dionysios Neofytos
- Division of Infectious Diseases, Transplant Infectious Disease Service, University Hospital of Geneva, Geneva, Switzerland
| | - Matteo Oliverio
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Translational Research, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne, Germany
| | - Juergen Prattes
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Translational Research, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne, Germany; University of Cologne, Faculty of Medicine and University Hospital Cologne, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD) and Excellence Center for Medical Mycology (ECMM), Cologne, Germany; German Centre for Infection Research (DZIF), Partner Site Bonn-Cologne, Cologne, Germany; Medical University of Graz, Department of Internal Medicine, Division of Infectious Disease, Excellence Center for Medical Mycology (ECMM), Graz, Austria
| | - Ilana Reinhold
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital of Zurich, Zurich, Switzerland
| | - Oliver A Cornely
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Translational Research, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne, Germany; University of Cologne, Faculty of Medicine and University Hospital Cologne, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD) and Excellence Center for Medical Mycology (ECMM), Cologne, Germany; German Centre for Infection Research (DZIF), Partner Site Bonn-Cologne, Cologne, Germany; University of Cologne, Faculty of Medicine and University Hospital Cologne, Clinical Trials Centre Cologne (ZKS Köln), Cologne, Germany
| | - Jannik Stemler
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Translational Research, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne, Germany; University of Cologne, Faculty of Medicine and University Hospital Cologne, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD) and Excellence Center for Medical Mycology (ECMM), Cologne, Germany; German Centre for Infection Research (DZIF), Partner Site Bonn-Cologne, Cologne, Germany.
| |
Collapse
|
12
|
Ishikawa K, Masaki T, Kawai F, Ota E, Mori N. Systematic Review of the Short-Term versus Long-Term Duration of Antibiotic Management for Neutropenic Fever in Patients with Cancer. Cancers (Basel) 2023; 15:1611. [PMID: 36900403 PMCID: PMC10001032 DOI: 10.3390/cancers15051611] [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: 01/24/2023] [Revised: 02/23/2023] [Accepted: 03/02/2023] [Indexed: 03/08/2023] Open
Abstract
Early antibiotic discontinuation has been proposed in patients with hematologic malignancy with fever of unknown origin during febrile neutropenia (FN). We intended to investigate the safety of early antibiotic discontinuation in FN. Two reviewers independently searched for articles from Embase, CENTRAL, and MEDLINE on 30 September 2022. The selection criteria were randomized control trials (RCTs) comparing short- and long-term durations for FN in cancer patients, and evaluating mortality, clinical failure, and bacteremia. Risk ratios (RRs) with 95% confidence intervals (CIs) were calculated. We identified eleven RCTs (comprising 1128 distinct patients with FN) from 1977 to 2022. A low certainty of evidence was observed, and no significant differences in mortality (RR 1.43, 95% CI, 0.81, 2.53, I2 = 0), clinical failure (RR 1.14, 95% CI, 0.86, 1.49, I2 = 25), or bacteremia (RR 1.32, 95% CI, 0.87, 2.01, I2 = 34) were identified, indicating that the efficacy of short-term treatment may not differ statistically from that of long-term treatment. Regarding patients with FN, our findings provide weak conclusions regarding the safety and efficacy of antimicrobial discontinuation prior to neutropenia resolution.
Collapse
Affiliation(s)
- Kazuhiro Ishikawa
- Department of Infectious Diseases, St. Luke’s International Hospital, Tokyo 104-8560, Japan
| | - Tetsuhiro Masaki
- Department of Infectious Diseases, St. Luke’s International Hospital, Tokyo 104-8560, Japan
| | - Fujimi Kawai
- Library, Center for Academic Resources, St. Luke’s International University, Tokyo 104-0044, Japan
| | - Erika Ota
- Global Health Nursing, Graduate School of Nursing Sciences, St. Luke’s International University, Tokyo 104-0044, Japan
- Tokyo Foundation for Policy Research, Tokyo 106-6234, Japan
| | - Nobuyoshi Mori
- Department of Infectious Diseases, St. Luke’s International Hospital, Tokyo 104-8560, Japan
| |
Collapse
|
13
|
Bochennek K, Hogardt M, Lehrnbecher T. Immune signatures, testing, and management of febrile neutropenia in pediatric cancer patients. Expert Rev Clin Immunol 2023; 19:267-277. [PMID: 36635981 DOI: 10.1080/1744666x.2023.2168646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
INTRODUCTION Infectious complications, particularly invasive bacterial and fungal infections, are still a major cause of morbidity in pediatric cancer patients and are associated with significant mortality. Over the last few years, there has been much effort in defining risk groups to tailor antimicrobial therapy, and in establishing pediatric-specific guidelines for antimicrobial strategies. AREAS COVERED This review provides a critical overview of defining risk groups for infection, diagnostic work-up, antimicrobial prophylaxis, empirical therapy, and treatment of established infections. EXPERT OPINION To date, no generalizable risk prediction model has been established for pediatric cancer patients. There is growing interest in defining the impact of the individual genetic background on infectious complications. New diagnostic tools have been developed over the last few years, but they need to be validated in pediatric cancer patients. International, pediatric-specific guidelines for antimicrobial prophylaxis, empirical therapy, and treatment of established infections have recently been published and will harmonize antimicrobial strategies in the future.
Collapse
Affiliation(s)
- Konrad Bochennek
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, University Hospital, Goethe University Frankfurt am Main, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany
| | - Michael Hogardt
- Institute of Medical Microbiology and Infection Control, University Hospital, Goethe University Frankfurt am Main, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany
| | - Thomas Lehrnbecher
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, University Hospital, Goethe University Frankfurt am Main, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany
| |
Collapse
|
14
|
Maertens J, Lodewyck T, Donnelly JP, Chantepie S, Robin C, Blijlevens N, Turlure P, Selleslag D, Baron F, Aoun M, Heinz WJ, Bertz H, Ráčil Z, Vandercam B, Drgona L, Coiteux V, Llorente CC, Schaefer-Prokop C, Paesmans M, Ameye L, Meert L, Cheung KJ, Hepler DA, Loeffler J, Barnes R, Marchetti O, Verweij P, Lamoth F, Bochud PY, Schwarzinger M, Cordonnier C, for the Infectious Diseases Group and the Acute Leukemia Group of the European Organization for Research and Treatment of Cancer. Empiric vs Preemptive Antifungal Strategy in High-Risk Neutropenic Patients on Fluconazole Prophylaxis: A Randomized Trial of the European Organization for Research and Treatment of Cancer. Clin Infect Dis 2023; 76:674-682. [PMID: 35906831 PMCID: PMC9938744 DOI: 10.1093/cid/ciac623] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 07/12/2022] [Accepted: 07/27/2022] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND Empiric antifungal therapy is considered the standard of care for high-risk neutropenic patients with persistent fever. The impact of a preemptive, diagnostic-driven approach based on galactomannan screening and chest computed tomography scan on demand on survival and on the risk of invasive fungal disease (IFD) during the first weeks of high-risk neutropenia is unknown. METHODS Patients with acute myeloid leukemia (AML) or myelodysplastic syndrome (MDS) and allogeneic hematopoietic cell transplant recipients were randomly assigned to receive caspofungin empirically (arm A) or preemptively (arm B), while receiving fluconazole 400 mg daily prophylactically. The primary end point of this noninferiority study was overall survival (OS) 42 days after randomization. RESULTS Of 556 patients recruited, 549 were eligible: 275 in arm A and 274 in arm B. Eighty percent of the patients had AML or MDS requiring high-dose chemotherapy, and 93% of them were in the first induction phase. At day 42, the OS was not inferior in arm B (96.7%; 95% confidence interval [CI], 93.8%-98.3%) when compared with arm A (93.1%; 95% CI, 89.3%-95.5%). The rates of IFDs at day 84 were not significantly different, 7.7% (95% CI, 4.5%-10.8%) in arm B vs 6.6% (95% CI, 3.6%-9.5%) in arm A. The rate of patients who received caspofungin was significantly lower in arm B (27%) than in arm A (63%; P < .001). CONCLUSIONS The preemptive antifungal strategy was safe for high-risk neutropenic patients given fluconazole as prophylaxis, halving the number of patients receiving antifungals without excess mortality or IFDs. Clinical Trials Registration. NCT01288378; EudraCT 2010-020814-27.
Collapse
Affiliation(s)
- Johan Maertens
- Correspondence: J. Maertens, University Hospitals Leuven, Herestraat 49, B-3000, Leuven, Belgium ()
| | - Tom Lodewyck
- Department of Hematology, Algemeen Ziekenhuis St Jan, Brugge, Belgium
| | - J Peter Donnelly
- Department of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
| | | | - Christine Robin
- Department of Hematology, Centre Hospitalier Universitaire Henri Mondor, Créteil, France
| | - Nicole Blijlevens
- Department of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Pascal Turlure
- Department of Hematology, Centre Hospitalier Universitaire Limoges, Limoges, France
| | - Dominik Selleslag
- Department of Hematology, Algemeen Ziekenhuis St Jan, Brugge, Belgium
| | - Frédéric Baron
- Department of Hematology, University of Liège and University Hospital of Liège, Liège, Belgium
| | - Mickael Aoun
- Department of Internal Medicine, Institut Jules Bordet, Brussels, Belgium
| | - Werner J Heinz
- Department of Hematology/Oncology, Caritas Hospital, Bad Mergentheim, Germany
| | - Hartmut Bertz
- Department of Hematology/Oncology, Faculty of Medicine and Medical Centre, University of Freiburg, Freiburg, Germany
| | - Zdeněk Ráčil
- Department of Hematology, Masaryk University Brno and Institute of Hematology and Blood Transfusion, Prague, Czech Republic
| | - Bernard Vandercam
- Department of Internal Medicine/Infectious Diseases, Cliniques Universitaires St. Luc, Brussels, Belgium
| | - Lubos Drgona
- Department of Oncohematology, Comenius University and National Cancer Institute, Bratislava, Slovakia
| | - Valerie Coiteux
- Service des maladies du sang, Centre Hospitalier Régional Universitaire Lille, Lille, France
| | | | | | - Marianne Paesmans
- Department of Internal Medicine, Institut Jules Bordet, Brussels, Belgium
| | - Lieveke Ameye
- Department of Internal Medicine, Institut Jules Bordet, Brussels, Belgium
| | - Liv Meert
- European Organisation for Research and Treatment of Cancer Headquarters, Brussels, Belgium
| | - Kin Jip Cheung
- European Organisation for Research and Treatment of Cancer Headquarters, Brussels, Belgium
| | | | - Jürgen Loeffler
- Department of Internal Medicine II, Universitaetsklinikum, Würzburg, Germany
| | - Rosemary Barnes
- Department of Infection, Immunity and Biochemistry, Cardiff University, Cardiff, United Kingdom
| | - Oscar Marchetti
- Department of Medicine, Lausanne University Hospital, Lausanne, Switzerland
- Department of Infectious Diseases, Ensemble Hospitalier de la Côte, Morges, Switzerland
| | - Paul Verweij
- Department of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Frederic Lamoth
- Department of Medicine, Lausanne University Hospital, Lausanne, Switzerland
| | - Pierre-Yves Bochud
- Department of Medicine, Lausanne University Hospital, Lausanne, Switzerland
| | - Michael Schwarzinger
- Translational Health Economics Network, Bordeaux University Hospital, Bordeaux, France
| | - Catherine Cordonnier
- Department of Hematology, Centre Hospitalier Universitaire Henri Mondor, Créteil, France
| | | |
Collapse
|
15
|
Efficacy and safety of echinocandin monotherapy and combination therapy for immunocompromised patients with systemic candidiasis: A systematic review and meta-analysis. J Mycol Med 2023; 33:101362. [PMID: 36867970 DOI: 10.1016/j.mycmed.2023.101362] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 02/10/2023] [Accepted: 02/10/2023] [Indexed: 02/17/2023]
Abstract
BACKGROUND Systemic candidiasis is caused by Candida invading the bloodstream. The efficacy and safety of echinocandins in monotherapy and combination therapy regimes have not been adequately compared in immunocompromised patients with Candidiasis, and thus this systematic review aims to do so. METHODS A protocol was prepared a priori. PubMed, Embase and Cochrane Library databases were searched systematically (from inception of each database to September 2022) to identify randomized controlled trials. Two reviewers performed screening, quality assessment of trials, and extracted data independently. Pairwise meta-analysis was performed using random-effects model to compare echinocandin monotherapy versus other antifungals. The primary outcomes of interest were treatment success and treatment-related adverse events. RESULTS 547 records (PubMed=310, EMBASE=210 and Cochrane Library=27) were reviewed. Following our screening criteria, six trials involving 177 patients were included. Risk of bias of four included studies had some concerns due to lack of a pre-specified analysis plan. Meta-analysis shows that echinocandin monotherapy does not have significantly higher rates of "treatment success" compared to other classes of antifungals (RR 1.12, 95%CI 0.80-1.56). However, echinocandins appeared to be significantly safer than other forms of antifungal therapy (RR 0.79, 95%CI 0.73-0.86). CONCLUSION Our findings have shown that echinocandin monotherapy (micafungin, caspofungin) given intravenously are just as effective as other antifungals (amphotericin B, itraconazole) in the treatment of systemic candidiasis in immunocompromised patients. There appears to be similar benefits when using echinocandins compared to amphotericin B which has also been used as a broad-spectrum antifungal, while avoiding the severe adverse effects that amphotericin B causes, such as nephrotoxicity.
Collapse
|
16
|
Singla RK, De R, Efferth T, Mezzetti B, Sahab Uddin M, Sanusi, Ntie-Kang F, Wang D, Schultz F, Kharat KR, Devkota HP, Battino M, Sur D, Lordan R, Patnaik SS, Tsagkaris C, Sai CS, Tripathi SK, Găman MA, Ahmed MEO, González-Burgos E, Babiaka SB, Paswan SK, Odimegwu JI, Akram F, Simal-Gandara J, Urquiza MS, Tikhonov A, Mondal H, Singla S, Lonardo SD, Mulholland EJ, Cenanovic M, Maigoro AY, Giampieri F, Lee S, Tzvetkov NT, Louka AM, Verma P, Chopra H, Olea SP, Khan J, Alvarez Suarez JM, Zheng X, Tomczyk M, Sabnani MK, Medina CDV, Khalid GM, Boyina HK, Georgiev MI, Supuran CT, Sobarzo-Sánchez E, Fan TP, Pittala V, Sureda A, Braidy N, Russo GL, Vacca RA, Banach M, Lizard G, Zarrouk A, Hammami S, Orhan IE, Aggarwal BB, Perry G, Miller MJ, Heinrich M, Bishayee A, Kijjoa A, Arkells N, Bredt D, Wink M, Fiebich BL, Kiran G, Yeung AWK, Gupta GK, Santini A, Lucarini M, Durazzo A, El-Demerdash A, Dinkova-Kostova AT, Cifuentes A, Souto EB, Zubair MAM, Badhe P, Echeverría J, Horbańczuk JO, Horbanczuk OK, Sheridan H, Sheshe SM, Witkowska AM, Abu-Reidah IM, Riaz M, Ullah H, Oladipupo AR, Lopez V, Sethiya NK, Shrestha BG, Ravanan P, Gupta SC, et alSingla RK, De R, Efferth T, Mezzetti B, Sahab Uddin M, Sanusi, Ntie-Kang F, Wang D, Schultz F, Kharat KR, Devkota HP, Battino M, Sur D, Lordan R, Patnaik SS, Tsagkaris C, Sai CS, Tripathi SK, Găman MA, Ahmed MEO, González-Burgos E, Babiaka SB, Paswan SK, Odimegwu JI, Akram F, Simal-Gandara J, Urquiza MS, Tikhonov A, Mondal H, Singla S, Lonardo SD, Mulholland EJ, Cenanovic M, Maigoro AY, Giampieri F, Lee S, Tzvetkov NT, Louka AM, Verma P, Chopra H, Olea SP, Khan J, Alvarez Suarez JM, Zheng X, Tomczyk M, Sabnani MK, Medina CDV, Khalid GM, Boyina HK, Georgiev MI, Supuran CT, Sobarzo-Sánchez E, Fan TP, Pittala V, Sureda A, Braidy N, Russo GL, Vacca RA, Banach M, Lizard G, Zarrouk A, Hammami S, Orhan IE, Aggarwal BB, Perry G, Miller MJ, Heinrich M, Bishayee A, Kijjoa A, Arkells N, Bredt D, Wink M, Fiebich BL, Kiran G, Yeung AWK, Gupta GK, Santini A, Lucarini M, Durazzo A, El-Demerdash A, Dinkova-Kostova AT, Cifuentes A, Souto EB, Zubair MAM, Badhe P, Echeverría J, Horbańczuk JO, Horbanczuk OK, Sheridan H, Sheshe SM, Witkowska AM, Abu-Reidah IM, Riaz M, Ullah H, Oladipupo AR, Lopez V, Sethiya NK, Shrestha BG, Ravanan P, Gupta SC, Alzahrani QE, Dama Sreedhar P, Xiao J, Moosavi MA, Subramani PA, Singh AK, Chettupalli AK, Patra JK, Singh G, Karpiński TM, Al-Rimawi F, Abiri R, Ahmed AF, Barreca D, Vats S, Amrani S, Fimognari C, Mocan A, Hritcu L, Semwal P, Shiblur Rahaman M, Emerald M, Akinrinde AS, Singh A, Joshi A, Joshi T, Khan SY, Balla GOA, Lu A, Pai SR, Ghzaiel I, Acar N, Es-Safi NE, Zengin G, Kureshi AA, Sharma AK, Baral B, Rani N, Jeandet P, Gulati M, Kapoor B, Mohanta YK, Emam-Djomeh Z, Onuku R, Depew JR, Atrooz OM, Goh BH, Andrade JC, Konwar B, Shine VJ, Ferreira JMLD, Ahmad J, Chaturvedi VK, Skalicka-Woźniak K, Sharma R, Gautam RK, Granica S, Parisi S, Kumar R, Atanasov AG, Shen B. The International Natural Product Sciences Taskforce (INPST) and the power of Twitter networking exemplified through #INPST hashtag analysis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 108:154520. [PMID: 36334386 DOI: 10.1016/j.phymed.2022.154520] [Show More Authors] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 07/12/2022] [Accepted: 10/21/2022] [Indexed: 06/16/2023]
Abstract
BACKGROUND The development of digital technologies and the evolution of open innovation approaches have enabled the creation of diverse virtual organizations and enterprises coordinating their activities primarily online. The open innovation platform titled "International Natural Product Sciences Taskforce" (INPST) was established in 2018, to bring together in collaborative environment individuals and organizations interested in natural product scientific research, and to empower their interactions by using digital communication tools. METHODS In this work, we present a general overview of INPST activities and showcase the specific use of Twitter as a powerful networking tool that was used to host a one-week "2021 INPST Twitter Networking Event" (spanning from 31st May 2021 to 6th June 2021) based on the application of the Twitter hashtag #INPST. RESULTS AND CONCLUSION The use of this hashtag during the networking event period was analyzed with Symplur Signals (https://www.symplur.com/), revealing a total of 6,036 tweets, shared by 686 users, which generated a total of 65,004,773 impressions (views of the respective tweets). This networking event's achieved high visibility and participation rate showcases a convincing example of how this social media platform can be used as a highly effective tool to host virtual Twitter-based international biomedical research events.
Collapse
Affiliation(s)
- Rajeev K Singla
- Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Xinchuan Road 2222, Chengdu, Sichuan, China; School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab-144411, India
| | - Ronita De
- ICMR-National Institute of Cholera and Enteric Diseases, P-33, CIT Rd, Subhas Sarobar Park, Phool Bagan, Beleghata, Kolkata, West Bengal 700010, India
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Mainz, Germany
| | - Bruno Mezzetti
- Department of Agriculture, Food and Environmental Sciences (D3A) Università Politecnica Delle Marche Ancona, IT, Italy
| | - Md Sahab Uddin
- Department of Pharmacy, Southeast University, Dhaka, Bangladesh; Pharmakon Neuroscience Research Network, Dhaka, Bangladesh
| | - Sanusi
- Research Center for Population - Indonesian Institute of Sciences, Indonesia
| | - Fidele Ntie-Kang
- Department of Chemistry, Faculty of Science, University of Buea, Buea P.O. Box 63, Cameroon
| | - Dongdong Wang
- Centre for Metabolism, Obesity, and Diabetes Research, Department of Medicine, McMaster University, HSC 4N71, 1280 Main Street West, Hamilton, ON L8S 4K1, Canada
| | - Fabien Schultz
- Technical University of Berlin, Institute of Biotechnology, Faculty III - Process Sciences, Gustav-Meyer-Allee 25, Berlin 13355, Germany; Neubrandenburg University of Applied Sciences, Department of Agriculture and Food Sciences, Brodaer Str. 2, Neubrandenburg 17033, Germany
| | | | - Hari Prasad Devkota
- Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1Oe-honmachi, Kumamoto 862-0973, Japan; Program for Leading Graduate Schools, HIGO Program, Kumamoto University, Japan
| | - Maurizio Battino
- Department of Clinical Sciences, Faculty of Medicine, Polytechnic University of Marche, Ancona 60131, Italy; International Research Center for Food Nutrition and Safety, Jiangsu University, Zhenjiang 212013, China
| | - Daniel Sur
- Department of Medical Oncology, "Iuliu Hatieganu" University of Medicine and Pharmacy Cluj-Napoca, Romania
| | - Ronan Lordan
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, PA, United States
| | - Sourav S Patnaik
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, United States
| | | | - Chandragiri Siva Sai
- Amity Institute of Pharmacy, Amity University, Uttar Pradesh, Lucknow Campus, Gomati Nagar, Lucknow, Uttar Pradesh 226010, India
| | - Surya Kant Tripathi
- Cancer Drug Resistance Laboratory, National Institute of Technology Rourkela, Odisha-769008, India
| | - Mihnea-Alexandru Găman
- ″Carol Davila" University of Medicine and Pharmacy, 8 Eroii Sanitari Boulevard, Bucharest, Romania; Center of Hematology and Bone Marrow Transplantation, Fundeni Clinical Institute, 258 Fundeni Road, Bucharest, Romania
| | - Mosa E O Ahmed
- Department of Pharmacognosy, Faculty of Pharmacy, Al Neelain University, Khartoum, Sudan
| | - Elena González-Burgos
- Department of Pharmacology, Pharmacognosy and Botany, University Complutense of Madrid, Spain
| | - Smith B Babiaka
- Department of Chemistry, Faculty of Science, University of Buea, Buea P.O. Box 63, Cameroon
| | | | | | - Faizan Akram
- Bahawalpur College of Pharmacy (BCP), Bahawalpur Medical and Dental College (BMDC), Bahawalpur, Pakistan
| | - Jesus Simal-Gandara
- Universidade de Vigo, Nutrition and Bromatology Group, Department of Analytical Chemistry and Food Science, Faculty of Science, Ourense E-32004, Spain
| | | | - Aleksei Tikhonov
- Translational Research Laboratory in Immunotherapy, Gustave Roussy, Villejuif, France
| | - Himel Mondal
- Department of Physiology, All India Institute of Medical Sciences, Deoghar, Jharkhand, India
| | - Shailja Singla
- iGlobal Research and Publishing Foundation, New Delhi, India
| | - Sara Di Lonardo
- Research Institute on Terrestrial Ecosystems-Italian National Research Council (IRET-CNR), Via Madonna del Piano 10, Sesto Fiorentino Fi 50019, Italy
| | - Eoghan J Mulholland
- Gastrointestinal Stem Cell Biology Laboratory, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom; Somerville College, University of Oxford, Oxford, United Kingdom
| | | | | | - Francesca Giampieri
- Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, Jeddah, Saudi Arabia; Research Group on Food, Nutritional Biochemistry and Health, Universidad Europea del Atlántico, Santander, Spain
| | - Soojin Lee
- Department of Bioscience and Biotechnology, Chungnam National University, Republic of Korea
| | - Nikolay T Tzvetkov
- Department of Biochemical Pharmacology and Drug Design, Institute of Molecular Biology "Roumen Tsanev", Bulgarian Academy of Sciences, Bulgaria
| | | | - Pritt Verma
- Department of Pharmacology, CSIR-NBRI, Lucknow, India
| | - Hitesh Chopra
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | | | - Johra Khan
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Al Majmaah 11952, Saudi Arabia
| | - José M Alvarez Suarez
- Departamento de Ingeniería en Alimentos, Colegio de Ciencias e Ingenierías, Universidad San Francisco de Quito, Quito, Ecuador
| | - Xiaonan Zheng
- Department of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Michał Tomczyk
- Department of Pharmacognosy, Faculty of Pharmacy with the Division of Laboratory Medicine, Medical University of Białystok, ul. Mickiewicza 2a, Białystok 15-230, Poland
| | - Manoj Kumar Sabnani
- The University of Texas at Arlington, United States; Alloy Therapeutics, United States
| | | | - Garba M Khalid
- Pharmaceutical Engineering Group, School of Pharmacy, Queen's University, Belfast BT9, United Kingdom
| | - Hemanth Kumar Boyina
- School of Pharmacy, Department of Pharmacology, Anurag University, Venkatapur, Medchal, Hyderabad, Telangana 500088, India
| | - Milen I Georgiev
- Laboratory of Metabolomics, Stephan Angeloff Institute of Microbiology, Bulgarian Academy of Sciences, 139 Ruski Blvd., Plovdiv 4000, Bulgaria
| | | | - Eduardo Sobarzo-Sánchez
- Instituto de Investigación y Postgrado, Facultad de Ciencias de la Salud, Universidad Central de Chile, Santiago 8330507, Chile; Department of Organic Chemistry, Faculty of Pharmacy, University of Santiago de Compostela, Santiago de Compostela 15782, Spain
| | - Tai-Ping Fan
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Science and Medicine, Northwest University, Xi'an, China
| | - Valeria Pittala
- Department of Drug and Health Sciences, University of Catania, Catania, Italy
| | - Antoni Sureda
- Research Group in Community Nutrition and Oxidative Stress, University of the Balearic Islands-IUNICS, Health Research Institute of Balearic Islands (IdISBa), and CIBEROBN (Physiopathology of Obesity and Nutrition), Palma, Balearic Islands E-07122, Spain
| | - Nady Braidy
- Centre for Healthy Brain Ageing (CHeBA), School of Psychiatry, University of New South Wales, Sydney, Australia
| | - Gian Luigi Russo
- National Research Council, Institute of Food Sciences, Avellino 83100, Italy
| | - Rosa Anna Vacca
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, National Council of Research, Bari 70126, Italy
| | - Maciej Banach
- Department of Preventive Cardiology and Lipidology, Medical University of Lodz (MUL), Lodz, Poland; Cardiovascular Research Centre, University of Zielona Gora, Zielona Gora, Poland
| | - Gérard Lizard
- Université de Bourgogne / Inserm, Laboratoire Bio-PeroxIL, Faculté des Sciences Gabriel, 6 Boulevard Gabriel, Dijon 21000 France
| | - Amira Zarrouk
- University of Monastir (Tunisia), Faculty of Medicine, LR-NAFS 'Nutrition - Functional Food & Vascular Health', Tunisia
| | - Sonia Hammami
- University of Monastir (Tunisia), Faculty of Medicine, LR-NAFS 'Nutrition - Functional Food & Vascular Health', Tunisia
| | - Ilkay Erdogan Orhan
- Department of Pharmacognosy, Faculty of Pharmacy, Gazi University, Ankara 06330, Türkiye
| | | | - George Perry
- Department of Neuroscience, Developmental, and Regenerative Biology, University of Texas, United States
| | | | | | - Anupam Bishayee
- College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, FL 34211, United States
| | - Anake Kijjoa
- Instituto de Ciências Biomédicas Abel Salazar e CIIMAR, Universidade do Porto, Portugal
| | - Nicolas Arkells
- International Natural Product Sciences Taskforce (INSPT), United States
| | | | - Michael Wink
- Heidelberg University, Institute of Pharmacy and Molecular Biotechnology (IPMB), Heidelberg 69120, Germany
| | - Bernd L Fiebich
- Neurochemistry and Neuroimmunology Research Group, Department of Psychiatry and Psychotherapy, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | | | - Andy Wai Kan Yeung
- Oral and Maxillofacial Radiology, Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, University of Hong Kong, Hong Kong, China
| | - Girish Kumar Gupta
- Department of Pharmaceutical Chemistry, Sri Sai College of Pharmacy, Badhani, Pathankot, Punjab, India
| | - Antonello Santini
- University of Napoli Federico II, Department of Pharmacy. Via D Montesano 49, Napoli 80131, Italy
| | - Massimo Lucarini
- CREA-Research Centre for Food and Nutrition, Via Ardeatina 546 00178 Rome, Italy
| | - Alessandra Durazzo
- CREA-Research Centre for Food and Nutrition, Via Ardeatina 546 00178 Rome, Italy
| | - Amr El-Demerdash
- Metabolic Biology & Biological Chemistry Department, John Innes Centre, Norwich Research Park, Norwich NR4 7UH, United Kingdom; Organic Chemistry Division, Chemistry Department, Faculty of Science, Mansoura University, Mansoura 35516, Egypt
| | | | | | - Eliana B Souto
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, Porto 4050-313, Portugal
| | | | - Pravin Badhe
- Swalife Foundation, India; Swalife Biotech Ltd, Ireland; Sinhgad College of Pharmacy, Vadgaon (BK) Pune Maharashtra India
| | - Javier Echeverría
- Departamento de Ciencias del Ambiente, Facultad de Química y Biología, Universidad de Santiago de Chile, Chile
| | - Jarosław Olav Horbańczuk
- Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences, Jastrzębiec 05-552, Poland
| | - Olaf K Horbanczuk
- Department of Technique and Food Product Development, Warsaw University of Life Sciences (WULS-SGGW) 159c Nowoursynowska, Warsaw 02-776, Poland
| | - Helen Sheridan
- The NatPro Centre. Trinity College Dublin. Dublin 2, Ireland
| | | | | | - Ibrahim M Abu-Reidah
- School of Science and the Environment, Grenfell Campus, Memorial University of Newfoundland, Corner Brook A2H 5G4, Canada
| | - Muhammad Riaz
- Department of Pharmacy, Shaheed Benazir Bhutto University, Sheringal 18050, Pakistan
| | - Hammad Ullah
- Department of Pharmacy, University of Naples Federico II, Naples 80131, Italy
| | - Akolade R Oladipupo
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Lagos, Nigeria; Department of Chemistry, Nelson Mandela University, Port Elizabeth, South Africa
| | - Víctor Lopez
- Department of Pharmacy, Universidad San Jorge, Villanueva de Gállego (Zaragoza), Spain
| | | | | | - Palaniyandi Ravanan
- Department of Microbiology, School of Life Sciences, Central University of Tamil Nadu, Thiruvarur, India
| | - Subash Chandra Gupta
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi, India; Department of Biochemistry, All India Institute of Medical Sciences, Guwahati, Assam, India
| | - Qushmua E Alzahrani
- Department of Pharmacy/Nursing Medicine Health and Environment, University of the Region of Joinville (UNIVILLE) Brazil, Sana Catarina, Joinville, Brazil
| | | | | | - Mohammad Amin Moosavi
- Molecular Medicine Department, Institute of Medical Biotechnology, National Institute of Genetics Engineering and Biotechnology, Tehran P.O. Box: 14965/161, Iran
| | - Parasuraman Aiya Subramani
- Independent Researcher, Vels Institute of Science, Technology and Advanced Studies (VISTAS), Chennai, India - 600048. formerly, Pallavaram, Chennai 600117, India
| | - Amit Kumar Singh
- Department of Biochemistry, University of Allahabad, Prayagraj 211002 India
| | | | - Jayanta Kumar Patra
- Research Institute of Integrative Life Sciences, Dongguk University-Seoul, Goyangsi 10326, Republic of Korea
| | - Gopal Singh
- Department of Plant Functional Metabolomics, Institute of Bioorganic Chemistry, Polish Academy of Sciences, Poznan, Poland
| | - Tomasz M Karpiński
- Chair and Department of Medical Microbiology, Poznań University of Medical Sciences, Wieniawskiego 3, Poznań 61-712, Poland
| | | | - Rambod Abiri
- Department of Forestry Science and Biodiversity, Faculty of Forestry and Environment, Universiti Putra Malaysia, UPM Serdang, Selangor 43400, Malaysia
| | - Atallah F Ahmed
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; Department of Pharmacognosy, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| | - Davide Barreca
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, Università degli Studi di Messina, Messina, Italy
| | - Sharad Vats
- Department of Bioscience and Biotechnology, Banasthali Vidyapith, Rajasthan 304022, India
| | - Said Amrani
- Laboratoire de Biologie et de Physiologie des Organismes, Faculté des Sciences Biologiques, USTHB, Bab Ezzouar, Alger, Algeria
| | | | - Andrei Mocan
- Department of Pharmaceutical Botany, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Lucian Hritcu
- Department of Biology, Alexandru Ioan Cuza University of Iasi, Bd. Carol I, No. 11, Iasi 700506, Romania
| | - Prabhakar Semwal
- Department of Life Sciences, Graphic Era Deemed to be University, Dehradun, Uttarakhand 248002, India
| | - Md Shiblur Rahaman
- Department of Environmental and Preventive Medicine, Jichi Medical University School of Medicine, 3311-1 Yakushiji, Shimotsuke, Tochigi 329-0498, Japan
| | - Mila Emerald
- PHYTOCEUTICALS International™ & NOVOTEK Global Solutions™, Canada
| | - Akinleye Stephen Akinrinde
- Department of Veterinary Physiology and Biochemistry, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | | | - Ashima Joshi
- Sardar Bhagwan Singh University, Balawala, Dehradun, India
| | - Tanuj Joshi
- Department of Pharmaceutical Sciences, Bhimtal, Kumaun University (Nainital), India
| | - Shafaat Yar Khan
- Research Lab III, Hematology & Vascular Biology, Department of Zoology, University of Sargodha, Sargodha, Pakistan
| | - Gareeballah Osman Adam Balla
- Department of Pharmacology, College of Veterinary Medicine, Sudan University of Science and Technology, Hilat Kuku, Khartoum North P.O. Box No. 204, Sudan
| | - Aiping Lu
- School of Chinese Medicine, Hong Kong Baptist University, HongKong, China
| | - Sandeep Ramchandra Pai
- Department of Botany, Rayat Shikshan Sanstha's, Dada Patil Mahavidyalaya, Karjat, Maharashtra, India
| | - Imen Ghzaiel
- Université de Bourgogne, Inserm, Laboratoire Bio - PeroxIL, Faculté des Sciences Gabriel, 6 Boulevard Gabriel, Dijon 21000 France; University Tunis El Manar, Tunis, Tunisia
| | | | - Nour Eddine Es-Safi
- Mohammed V University in Rabat, LPCMIO, Materials Science Center (MSC), Ecole Normale Supérieure, Rabat, Morocco
| | - Gokhan Zengin
- Department of Biology, Science Faculty, Selcuk University, Konya, Turkey
| | - Azazahemad A Kureshi
- Department of Chemistry, Sardar Vallabhbhai National Institute of Technology, Surat, India
| | | | | | - Neeraj Rani
- Department of Pharmaceutical Sciences, Chaudhary Bansilal University, Bhiwani, Haryana, India
| | - Philippe Jeandet
- University of Reims, Research Unit Induced Resistance and Plant Bioprotection, USC INRAe 1488, Reims, France
| | - Monica Gulati
- School of Pharmaceutical Sciences, Lovely Professional University, Jalandhar-Delhi G.T. Road (NH 1) Phagwara, Punjab 144411 India
| | - Bhupinder Kapoor
- School of Pharmaceutical Sciences, Lovely Professional University, Jalandhar-Delhi G.T. Road (NH 1) Phagwara, Punjab 144411 India
| | - Yugal Kishore Mohanta
- Department of Applied Biology, School of Biological Sciences, University of Science and Technology Meghalaya (USTM), Techno City, Kling Road, Baridua, Ri-Bhoi, Meghalaya 793101, India
| | | | - Raphael Onuku
- Department of Pharmaceutical and Medicinal Chemistry, Faculty of Pharmaceutical Sciences, University of Nigeria, Nigeria
| | | | - Omar M Atrooz
- Department of Biological Sciences, Mutah University, Jordan
| | - Bey Hing Goh
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China; Biofunctional Molecule Exploratory (BMEX) Research Group, School of Pharmacy, Monash University Malaysia, Subang Jaya, Malaysia
| | - Jose Carlos Andrade
- TOXRUN - Toxicology Research Unit, University Institute of Health Sciences, CESPU, Gandra, Portugal
| | | | - V J Shine
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram, Kerala 695014, India
| | | | - Jamil Ahmad
- Department of Human Nutrition, The University of Agriculture Peshawar, Khyber Pakhtunkhwa, Pakistan
| | - Vivek K Chaturvedi
- Department of Gastroenterology, Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, India
| | | | - Rohit Sharma
- Department of Rasa Shastra and Bhaishajya Kalpana, Faculty of Ayurveda, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh 221005, India
| | - Rupesh K Gautam
- Deparment of Pharmacology, Indore Institute of Pharmacy, IIST Campus, Rau-Indore-453331, India
| | - Sebastian Granica
- Microbiota Lab, Department of Pharmacognosy and Molecular Basis of Phytotherapy, Medical University of Warsaw, Poland
| | - Salvatore Parisi
- Lourdes Matha Institute of Hotel Management and Catering Technology, Kerala State, India
| | - Rishabh Kumar
- School of Medical and Allied Sciences, K.R. Mangalam University, Sohna Road, Gurugram, Haryana 122103, India
| | - Atanas G Atanasov
- Ludwig Boltzmann Institute for Digital Health and Patient Safety, Medical University of Vienna, Spitalgasse 23, Vienna 1090, Austria; Department of Pharmaceutical Sciences, University of Vienna, Althanstraße 14, Vienna 1090, Austria; Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences, Jastrzebiec, Magdalenka 05-552, Poland.
| | - Bairong Shen
- Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Xinchuan Road 2222, Chengdu, Sichuan, China.
| |
Collapse
|
17
|
Maertens J, Pagano L, Azoulay E, Warris A. Liposomal amphotericin B-the present. J Antimicrob Chemother 2022; 77:ii11-ii20. [PMID: 36426672 PMCID: PMC9693760 DOI: 10.1093/jac/dkac352] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Most invasive fungal infections are opportunistic in nature but the epidemiology is constantly changing, with new risk groups being identified. Neutropenia is a classical risk factor for fungal infections, while critically ill patients in the ICU are now increasingly at risk of yeast and mould infections. Factors to be considered when choosing antifungal treatment include the emergence of rarer fungal pathogens, the risk of resistance to azoles and echinocandins and the possibility of drug-drug interactions. Liposomal amphotericin B has retained its place in the therapeutic armamentarium based on its clinical profile: a broad spectrum of antifungal activity with a low risk of resistance, predictable pharmacokinetics with a rapid accumulation at the infection site (including biofilms), a low potential for drug-drug interactions and a low risk of acute and chronic treatment-limiting toxicities versus other formulations of amphotericin B. It is a suitable choice for the first-line empirical or pre-emptive treatment of suspected fungal infections in neutropenic haematology patients and is an excellent alternative for patients with documented fungal disease who can no longer tolerate or continue their first-line azole or echinocandin therapy, both in the haematology setting and in the ICU. Moreover, it is the first-line drug of choice for the treatment of invasive mucormycosis. Finally, liposomal amphotericin B is one of the few antifungal agents approved for use in children of all ages over 1 month and is included in paediatric-specific guidelines for the management of fungal disease.
Collapse
Affiliation(s)
- J Maertens
- Department of Hematology, University Hospital Gasthuisberg, KU Leuven, Leuven, Belgium.,Department of Microbiology, Immunology, and Transplantation, KU Leuven, Leuven, Belgium
| | - L Pagano
- Sezione di Ematologia, Fondazione Policlinico Universitario Agostino Gemelli-IRCCS-Università Cattolica del Sacro Cuore, Rome, Italy
| | - E Azoulay
- Médecine Intensive et Réanimation, Hôpital Saint-Louis, APHP, University of Paris, Paris, France
| | - A Warris
- MRC Centre for Medical Mycology, University of Exeter, Exeter, UK.,Great Ormond Street Hospital, Paediatric Infectious Diseases Unit, London, UK
| |
Collapse
|
18
|
Itoh K, Shigemi H, Kinoshita K, Tsukasaki H, Imamura S, Morinaga K, Yoshio N, Nakayama T, Inoue H, Ueda T, Yamauchi T, Iwasaki H. Efficacy and Safety of Caspofungin Treatment in Febrile Neutropenic Patients with Hematological Disorders: A Multicenter Consecutive Case Series. Intern Med 2022; 61:3037-3044. [PMID: 35314551 PMCID: PMC9646351 DOI: 10.2169/internalmedicine.9070-21] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/05/2022] Open
Abstract
Introduction Invasive fungal infections have been attracting attention as significant fatal complications in patients with febrile neutropenia (FN) who undergo intensive chemotherapy or hematopoietic stem cell transplantation to treat hematological malignancies. Although clinical trials are already underway in other countries, evidence supporting the use of caspofungin (CAS) in FN patients in Japan is still insufficient. Methods A retrospective study of patients treated with CAS for FN associated with hematological diseases between April 2015 and March 2018 was conducted to determine the treatment efficacy and safety. The study was conducted as a multicenter collaboration, and the data of 52 patients who met all of the inclusion criteria were analyzed. A five-composite-endpoint method was used, and the treatment was judged to be effective when all five endpoints (defervescence during neutropenia; no breakthrough fungal infections; resolution of baseline fungal infections; a survival for seven days or more after the completion of therapy; and no discontinuation of therapy due to side effects or invalidity) were met. Results The efficacy rate was 53.8% (28/52), which is close to the average reported efficacy rate. Adverse events included liver dysfunction and electrolyte abnormalities, but no renal dysfunction or serious events were seen. Conclusion These results suggest that the use of CAS in FN patients with hematological diseases is effective and well-tolerated, and we believe that the use of CAS could become a significant treatment in Japan.
Collapse
Affiliation(s)
- Kazuhiro Itoh
- Department of Internal Medicine, National Hospital Organization Awara Hospital, Japan
| | - Hiroko Shigemi
- Department of Infection Control and Laboratory Medicine, Kyoto Prefectural University of Medicine, Japan
| | | | | | - Shin Imamura
- Department of Hematology, Red Cross Fukui Hospital, Japan
| | - Koji Morinaga
- Department of Hematology and Oncology, Fukui Prefectural Hospital, Japan
| | - Nobuyuki Yoshio
- Department of Hematology, National Hospital Organization Kanazawa Medical Center, Japan
| | - Takashi Nakayama
- Department of Oncology and Hematology, Fukui-ken Saiseikai Hospital, Japan
| | - Hitoshi Inoue
- Department of Internal Medicine, National Hospital Organization Tsuruga Medical Center, Japan
| | - Takanori Ueda
- Department of Hematology and Oncology, Faculty of Medical Sciences, University of Fukui, Japan
| | - Takahiro Yamauchi
- Department of Hematology and Oncology, Faculty of Medical Sciences, University of Fukui, Japan
| | - Hiromichi Iwasaki
- Department of Infection Control and Prevention, University of Fukui Hospital, Japan
| |
Collapse
|
19
|
Earnshaw SR, McDade C, Bryan A, Ines M, Micallef C, Sung A, Enoch DA. Real-World Financial and Clinical Impact of Diagnostic-Driven and Empirical-Treatment Strategies in High-Risk Immunocompromised Patients with Suspected Aspergillus Infection in the United Kingdom. Microbiol Spectr 2022; 10:e0042522. [PMID: 35532266 PMCID: PMC9241825 DOI: 10.1128/spectrum.00425-22] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 04/10/2022] [Indexed: 11/25/2022] Open
Abstract
A diagnostic-driven (DD) treatment strategy has proven successful for treating invasive fungal infections (IFIs) caused by Aspergillus. However, uptake of this treatment strategy is not fully embraced. This study compares the economic and clinical impact of DD and empirical-treatment (ET) strategies used within hospitals. Methods: a decision-analytic model was developed to compare costs and clinical outcomes associated with ET or a DD strategy of identifying infections caused by Aspergillus via galactomannan-antigen testing or Aspergillus polymerase chain reaction (PCR) in neutropenic patients with unexplained fever. Patients were treated prophylactically with antifungal treatments as seen in United Kingdom (UK) hospitals. The IFI incidence, response, mortality, resource use, and adverse events were obtained from meta-analyses and other clinical studies. Analyses were performed from the U.K. hospital perspective, and costs were obtained from standard costing sources. Although diagnostic-testing costs increased, total cost and length of stay were reduced by £1,121 and 1.54 days when treating via a DD strategy. Intensive care and general ward days accounted for > 40% of total costs and > 58% of the cost reduction came from reduced antifungal costs. Treating with a DD strategy reduced the number of patients being treated with antifungal agents while survival was increased. Thus, a DD strategy was cost savings (-£136,787 cost per death avoided) compared with an ET strategy. Conclusion: this study suggests that incorporating a DD strategy as the preferred treatment protocol may be a cost-saving and clinically improved treatment strategy for managing neutropenic patients with unexplained fever. IMPORTANCE Patients at risk of invasive fungal infections (IFIs), such as Aspergillus spp., tend to be immunocompromised and usually take several medications which may generate many side effects. Prescribing is further complicated by comorbidities, drug interactions and challenges accessing diagnostics. Therefore, adding another agent may be neither straightforward nor the best option for these types of patients. A diagnostic-driven (DD) treatment strategy has proven successful for treating IFIs. However, uptake of this treatment strategy is not fully embraced in clinical practice perhaps because this strategy is thought to be more costly and/or to result in higher mortality relative to treating empirically. We developed a decision-analytic model to examine the impact of these 2 strategies on costs and health outcomes. This study indicates that incorporating a DD strategy as the preferred treatment protocol may be a cost-saving and clinically improved treatment strategy for managing neutropenic patients with unexplained fever.
Collapse
Affiliation(s)
| | - Cheryl McDade
- RTI Health Solutions, Research Triangle Park, North Carolina, USA
| | - Andrew Bryan
- Pfizer Biopharmaceuticals Group, Pfizer Ltd., Surrey, United Kingdom
| | - Monica Ines
- Hospital & Vaccines Business Unit, Pfizer, Inc., Porto-Salvo, Portugal
| | | | | | - David A. Enoch
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| |
Collapse
|
20
|
Mujwar S, Tripathi A. Repurposing benzbromarone as antifolate to develop novel antifungal therapy for Candida albicans. J Mol Model 2022; 28:193. [PMID: 35716240 PMCID: PMC9206073 DOI: 10.1007/s00894-022-05185-w] [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: 11/08/2021] [Accepted: 06/07/2022] [Indexed: 12/23/2022]
Abstract
Fungal infections in humans are responsible for mild to severe infections resulting in systemic effects that cause a large amount of mortality. Invasive fungal infections are having similar symptomatic effects to those of COVID-19. The COVID-19 patients are immunocompromised in nature and have a high probability of developing severe fungal infections, resulting in the development of further complications. The existing antifungal therapy has associated problems related to the development of drug resistance, being sub-potent in nature, and the presence of undesirable toxic effects. The fungal dihydrofolate reductase is an essential enzyme involved in the absorption of dietary folic acid and its conversion into tetrahydrofolate, which is a coenzyme required for the biosynthesis of the fungal nucleotides. Thus, in the current study, an attempt has been made to identify potential folate inhibitors of Candida albicans by a computational drug repurposing approach. Based upon the molecular docking simulation-based virtual screening followed by the molecular dynamic simulation of the macromolecular complex, benzbromarone has been identified as a potential anti-folate agent for the development of a novel therapy for the treatment of candidiasis.
Collapse
Affiliation(s)
- Somdutt Mujwar
- M.M. College of Pharmacy, Maharishi Markandeshwar University, Mullana-133207 Haryana, India.
| | - Avanish Tripathi
- Institute of Pharmaceutical Research, GLA University, Mathura, 281406, Uttar Pradesh, India
| |
Collapse
|
21
|
DEVECİ B, YÜCEL OK, KUBLASHVİLİ G, ILTAR U, ATAŞ Ü, TOPTAS T, TURHAN Ö, SABA R. Akut Lenfoblastik Lösemili Hastalarda Mikafungin Profilaksisi. İSTANBUL GELIŞIM ÜNIVERSITESI SAĞLIK BILIMLERI DERGISI 2022. [DOI: 10.38079/igusabder.993158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
|
22
|
Fatahinia M, Ghojoghi A, Erfaninejad M, Ahmadpour E, Nazar E, Barac A. Non-albicans candidemia in cancer patients as an increasing health problem: A comprehensive review and meta-analysis. ASIAN PAC J TROP MED 2022. [DOI: 10.4103/1995-7645.356992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
23
|
Chau MM, Daveson K, Alffenaar JWC, Gwee A, Ho SA, Marriott DJE, Trubiano JA, Zhao J, Roberts JA. Consensus guidelines for optimising antifungal drug delivery and monitoring to avoid toxicity and improve outcomes in patients with haematological malignancy and haemopoietic stem cell transplant recipients, 2021. Intern Med J 2021; 51 Suppl 7:37-66. [PMID: 34937141 DOI: 10.1111/imj.15587] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Antifungal agents can have complex dosing and the potential for drug interaction, both of which can lead to subtherapeutic antifungal drug concentrations and poorer clinical outcomes for patients with haematological malignancy and haemopoietic stem cell transplant recipients. Antifungal agents can also be associated with significant toxicities when drug concentrations are too high. Suboptimal dosing can be minimised by clinical assessment, laboratory monitoring, avoidance of interacting drugs, and dose modification. Therapeutic drug monitoring (TDM) plays an increasingly important role in antifungal therapy, particularly for antifungal agents that have an established exposure-response relationship with either a narrow therapeutic window, large dose-exposure variability, cytochrome P450 gene polymorphism affecting drug metabolism, the presence of antifungal drug interactions or unexpected toxicity, and/or concerns for non-compliance or inadequate absorption of oral antifungals. These guidelines provide recommendations on antifungal drug monitoring and TDM-guided dosing adjustment for selected antifungal agents, and include suggested resources for identifying and analysing antifungal drug interactions. Recommended competencies for optimal interpretation of antifungal TDM and dose recommendations are also provided.
Collapse
Affiliation(s)
- Maggie M Chau
- Pharmacy Department, The Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Kathryn Daveson
- Department of Infectious Diseases and Microbiology, The Canberra Hospital, Garran, Australian Capital Territory, Australia
| | - Jan-Willem C Alffenaar
- Faculty of Medicine and Health, School of Pharmacy, University of Sydney, Camperdown, New South Wales, Australia.,Pharmacy Department, Westmead Hospital, Westmead, New South Wales, Australia.,Marie Bashir Institute of Infectious Diseases and Biosecurity, University of Sydney, Camperdown, New South Wales, Australia
| | - Amanda Gwee
- Infectious Diseases Unit, The Royal Children's Hospital, Parkville, Victoria, Australia.,Department of Paediatrics, The University of Melbourne, Parkville, Victoria, Australia.,Infectious Diseases Group, Murdoch Children's Research Institute, Parkville, Victoria, Australia
| | - Su Ann Ho
- Department of Infectious Diseases, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Deborah J E Marriott
- Department of Clinical Microbiology and Infectious Diseases, St Vincent's Hospital, Darlinghurst, New South Wales, Australia.,Faculty of Science, University of Technology, Ultimo, New South Wales, Australia.,Faculty of Medicine, The University of New South Wales, Kensington, New South Wales, Australia
| | - Jason A Trubiano
- Department of Infectious Diseases, Austin Health, Heidelberg, Victoria, Australia.,Department of Medicine, The University of Melbourne, Parkville, Victoria, Australia
| | - Jessie Zhao
- Department of Haematology, The Alfred Hospital, Melbourne, Victoria, Australia
| | - Jason A Roberts
- The University of Queensland Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia.,Department of Pharmacy and Intensive Care Medicine, Royal Brisbane and Women's Hospital, Herston, Queensland, Australia.,Division of Anaesthesiology Critical Care Emergency and Pain Medicine, Nîmes University Hospital, University of Montpellier, Nîmes, France
| | | |
Collapse
|
24
|
DeJarnette C, Meyer CJ, Jenner AR, Butts A, Peters T, Cheramie MN, Phelps GA, Vita NA, Loudon-Hossler VC, Lee RE, Palmer GE. Identification of Inhibitors of Fungal Fatty Acid Biosynthesis. ACS Infect Dis 2021; 7:3210-3223. [PMID: 34786940 DOI: 10.1021/acsinfecdis.1c00404] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Fungal fatty acid (FA) synthase and desaturase enzymes are essential for the growth and virulence of human fungal pathogens. These enzymes are structurally distinct from their mammalian counterparts, making them attractive targets for antifungal development. However, there has been little progress in identifying chemotypes that target fungal FA biosynthesis. To accomplish this, we applied a whole-cell-based method known as Target Abundance-based FItness Screening using Candida albicans. Strains with varying levels of FA synthase or desaturase expression were grown in competition to screen a custom small-molecule library. Hit compounds were defined as preferentially inhibiting the growth of the low target-expressing strains. Dose-response experiments confirmed that 16 hits (11 with an acyl hydrazide core) differentially inhibited the growth of strains with an altered desaturase expression, indicating a specific chemical-target interaction. Exogenous unsaturated FAs restored C. albicans growth in the presence of inhibitory concentrations of the most potent acyl hydrazides, further supporting the primary mechanism being inhibition of FA desaturase. A systematic analysis of the structure-activity relationship confirmed the acyl hydrazide core as essential for inhibitory activity. This collection demonstrated broad-spectrum activity against Candida auris and mucormycetes and retained the activity against azole-resistant candida isolates. Finally, a preliminary analysis of toxicity to mammalian cells identified potential lead compounds with desirable selectivities. Collectively, these results establish a scaffold that targets fungal FA biosynthesis with a potential for development into novel therapeutics.
Collapse
Affiliation(s)
- Christian DeJarnette
- Department of Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, Tennessee 38163, United States
| | - Chris J. Meyer
- Department of Chemical Biology and Therapeutics, St. Jude Children’s Research Hospital, Memphis, Tennessee 38105, United States
| | - Alexander R. Jenner
- Department of Chemical Biology and Therapeutics, St. Jude Children’s Research Hospital, Memphis, Tennessee 38105, United States
| | - Arielle Butts
- Department of Clinical Pharmacy and Translational Science, College of Pharmacy, University of Tennessee Health Sciences Center, Memphis, Tennessee 38163, United States
| | - Tracy Peters
- Department of Clinical Pharmacy and Translational Science, College of Pharmacy, University of Tennessee Health Sciences Center, Memphis, Tennessee 38163, United States
| | - Martin N. Cheramie
- Department of Chemical Biology and Therapeutics, St. Jude Children’s Research Hospital, Memphis, Tennessee 38105, United States
| | - Gregory A. Phelps
- Department of Chemical Biology and Therapeutics, St. Jude Children’s Research Hospital, Memphis, Tennessee 38105, United States
- Graduate School of Biomedical Sciences, St. Jude Children’s Research Hospital, Memphis Tennessee 38103, United States
| | - Nicole A. Vita
- Department of Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, Tennessee 38163, United States
| | - Victoria C. Loudon-Hossler
- Department of Chemical Biology and Therapeutics, St. Jude Children’s Research Hospital, Memphis, Tennessee 38105, United States
| | - Richard E. Lee
- Department of Chemical Biology and Therapeutics, St. Jude Children’s Research Hospital, Memphis, Tennessee 38105, United States
| | - Glen E. Palmer
- Department of Clinical Pharmacy and Translational Science, College of Pharmacy, University of Tennessee Health Sciences Center, Memphis, Tennessee 38163, United States
| |
Collapse
|
25
|
Yang YL, Xiang ZJ, Yang JH, Wang WJ, Xu ZC, Xiang RL. Adverse Effects Associated With Currently Commonly Used Antifungal Agents: A Network Meta-Analysis and Systematic Review. Front Pharmacol 2021; 12:697330. [PMID: 34776941 PMCID: PMC8585744 DOI: 10.3389/fphar.2021.697330] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 10/18/2021] [Indexed: 11/24/2022] Open
Abstract
Background: Invasive fungal infections (IFI) is an important contributing factor in morbidity and mortality of immunocompromised and critically ill patients. Although the therapeutic effects of these drugs on IFI have been well documented, the long-term use of antifungal agents has raised concerns about drug tolerability and treatment-related toxicity risks. Methods: We searched articles published before June 30, 2020 in four electronic databases: Web of Science, Cochrane Library, embase and PubMed. Results: 66 trials were determined to meet our inclusion criteria, providing data on 18,230 participants. We sorted out 23 AEs by system organ classes and six laboratory AEs, 13 of these were used to construct 13 network meta-analyses. Compared with LAmB, anidulafungin, caspofungin, micafungin, fluconazole, and posaconazole had a significantly low incidence of discontinuation of therapy due to AEs (OR = 0.24 (0.09,0.65), 0.24 (0.13,0.43), 0.32 (0.19,0.52), 0.38 (0.23,0.62) and 0.35 (0.17,0.69), respectively). Conclusion: We found that echinocandins are the most tolerated antifungal agents with high safety. The AEs of triazole drugs are mainly concentrated on the increase in liver enzymes, nervous system disorders, especially visual disorders, gastrointestinal disorders, and cardiac diseases. LAmB is the least tolerated and has the most abundant AEs.
Collapse
Affiliation(s)
- Yan-Li Yang
- Department of Pulmonary and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Zi-Jian Xiang
- Beijing Zhiyun Data Technology Co., Ltd., Beijing, China
| | - Jing-Hua Yang
- Beijing Zhiyun Data Technology Co., Ltd., Beijing, China
| | - Wen-Jie Wang
- Beijing Zhiyun Data Technology Co., Ltd., Beijing, China
| | - Zhi-Chun Xu
- Beijing Zhiyun Data Technology Co., Ltd., Beijing, China
| | - Ruo-Lan Xiang
- Department of Physiology and Pathophysiology, Peking University School of Basic Medical Sciences, Beijing, China
| |
Collapse
|
26
|
Methodological and reporting quality of non-inferiority randomized controlled trials comparing antifungal therapies: a systematic review. Clin Microbiol Infect 2021; 28:640-648. [PMID: 34763055 DOI: 10.1016/j.cmi.2021.11.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 10/30/2021] [Accepted: 11/01/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Detailed reporting is essential in non-inferiority randomized controlled trials (NI-RCTs) to assess evidence quality, as these trials inform standards of care. OBJECTIVES The primary objective was to evaluate the methodological and reporting quality of antifungal NI-RCTs. DATA SOURCES Medline, EMBASE, the Cochrane CENTRAL and the United States Federal Drug Administration (FDA) drugs database were searched to 9 September 2020. STUDY ELIGIBILITY CRITERIA NI-RCTs differing by antifungal formulation, type, dose, administration and/or duration were included. Articles were independently assessed in duplicate using quality indicators developed by the Consolidated Standards of Reporting Trials (CONSORT) group. PARTICIPANTS Patients enrolled in antifungal trials for prophylactic and therapeutic use. METHODS The Cochrane RoB 2.0 tool was used to assess risk of bias. Descriptive statistics were used; all statistical tests were two sided. RESULTS Of 32 included studies, 22 (68.7%) did not justify the NIM. Handling of missing data was not described in 20 (62.5%). Intention-to-treat (ITT) and per-protocol (PP) analyses were both reported in 12/32 (37.5%) studies. Eleven of 32 studies (34.3%) reported potentially misleading conclusions. Industry-financed studies were more likely to report only the ITT analysis (n = 14/27, 51.9%). Methodological and reporting quality was unaffected by publication year; risk of bias from missing data changed over time. Overall risk of bias across included studies was moderate to high, with high risk in randomization process (n = 8/32, 25%), missing outcome data (n = 5/32, 15.6%), and selection of reported result (n = 9/32, 28.1%). CONCLUSIONS Justification of the non-inferiority margin, reporting of ITT and PP analyses, missing data handling description, and ensuring conclusions are consistent with reported data is necessary to improve CONSORT adherence. Small sample size and overall risk of bias are study limitations. (Systematic Review Registration Number PROSPERO CRD42020219497).
Collapse
|
27
|
Are antifungal non-inferiority trials at risk of eroding effectiveness because of bio-creep? A secondary analysis of a systematic review. Antimicrob Agents Chemother 2021; 66:e0162721. [PMID: 34662190 DOI: 10.1128/aac.01627-21] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Non-inferiority randomized controlled trial (RCT) effectiveness may erode when results favour the active control over time, and when a decreasingly effective control arm is used in serial trials. We analyzed 32 antifungal noninferiority RCTs (NI-RCTs) for these scenarios in this secondary analysis of a systematic review. Our exploratory analysis suggests that the erosion risk in the effectiveness of antifungal non-inferiority trials is uncommon. Findings are limited by small sample size, and overall risk of bias.
Collapse
|
28
|
Beauchemin C, Guinan K, Claveau D, Dufresne SF, Rotstein C. Economic evaluation of isavuconazole for suspected invasive pulmonary aspergillosis in Canada. Expert Rev Pharmacoecon Outcomes Res 2021; 22:805-814. [PMID: 34524935 DOI: 10.1080/14737167.2021.1981862] [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] [Indexed: 10/20/2022]
Abstract
BACKGROUND Invasive mold infections (IMI) directly impact life expectancy, especially with delayed therapy. Among IMI, aspergillosis (IA) is more common than mucormycosis (IM), resulting in IA-targeted empirical treatment with voriconazole for suspected invasive pulmonary aspergillosis (IPA), despite IM ineffectiveness. Recently, isavuconazole was approved in Canada for IA and IM. The primary objective was to assess the cost-effectiveness of isavuconazole compared to voriconazole for suspected IPA in Canada. A secondary objective was to assess the impact of varying time horizons to address the wide spectrum of life expectancies, according to patients underlying diseases. RESEARCH DESIGN AND METHODS A 5-year decision-tree was developed from the Canadian Ministry of Health (MoH) and societal perspectives. Efficacy parameters were extracted from SECURE/VITAL trials. Costs included treatment acquisition, hospitalization, adverse events and productivity loss. 3- and 10-year time horizon alternative scenarios and extensive sensitivity analyses were also conducted. RESULTS From a MoH perspective, isavuconazole compared to voriconazole resulted in an incremental cost-utility ratio (ICUR) of $C30,160/QALY. 3- and10-year ICURs were also cost-effective, relative to a willingness-to-pay threshold of $C50,000/QALY. CONCLUSIONS This study demonstrates that, in comparison to voriconazole, isavuconazole is a cost-effective strategy for the treatment of patients with suspected IPA, regardless of their life expectancy.
Collapse
Affiliation(s)
- C Beauchemin
- Faculté de Pharmacie, Université de Montreal, Montreal, Canada.,PeriPharm Inc, Montreal, Canada
| | | | - D Claveau
- Medical Affairs, AVIR Pharma Inc, Blainville, Canada
| | - S Frédéric Dufresne
- Division of Infectious Diseases and Clinical Microbiology, Department of Medicine, Maisonneuve-Rosemont Hospital, Montreal, Canada.,Department of Microbiology, Immunology and Infectious Diseases, Faculty of Medicine, Université de Montréal, Montreal, Canada
| | - C Rotstein
- Immunocompromised Host Infectious Diseases Service, Division of Infectious Diseases, Department of Medicine, University Health Network, University of Toronto, Toronto, Canada
| |
Collapse
|
29
|
Saito AM, Yoshida I, Tanaka S, Sawamura M, Hidaka M, Yoshida S, Uike N, Kaneko Y, Miyazaki Y, Nagai H. Efficacy of Intravenous Itraconazole Versus Liposomal Amphotericin B as Empirical Antifungal Therapy in Hematological Malignancy with Persistent Fever and Neutropenia: Study Protocol for a Multicenter, Prospective, Randomized Non-inferiority Trial. Kurume Med J 2021; 66:239-246. [PMID: 34544939 DOI: 10.2739/kurumemedj.ms664001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Febrile neutropenia, a serious complication that can occur during the treatment of hematological malignancies, can sometimes be fatal owing to fungal infection. Prospective randomized trials indicated the utility of liposomal amphotericin B or caspofungin as an empirical antifungal therapy. Itraconazole, a broad-spectrum tri azole antifungal agent, is poorly absorbed in the intestines after oral absorption and makes it difficult to achieve a stable serum drug concentration. Therefore, an intravenous formulation might offer a potentially safer and more effective alternative. To compare the efficacy and safety of empirical antifungal therapy, patients will be randomly assigned to either the liposomal amphotericin B 3.0 mg/kg once daily group or the intravenous itraconazole 200 mg dose group with five stratification factors (disease risk, previous antifungal prophylaxis, age, sex, and institute). The primary endpoint will be overall favorable response, comprising five secondary endpoints: successful treatment of baseline infection by the end of the treatment; absence of breakthrough infection; no discontinuation of the antifungal treatment due to drug-related toxicity; fever resolution during neutropenia; and 7-day survival after termination of the antifungal treatment. The target sample size of 850 subjects is sufficient to prove the non inferiority of itraconazole compared with liposomal amphotericin B, with a non-inferiority margin of 10%, one sided significance level of 5%, and power of 90%.
Collapse
Affiliation(s)
- Akiko M Saito
- Laboratory of Clinical, Epidemiological and Health Services Research, Clinical Research Center, National Hospital Organization Nagoya Medical Center
| | - Isao Yoshida
- Department of Hematologic Oncology, National Hospital Organization Shikoku Cancer Center
| | - Shiro Tanaka
- Department of Clinical Biostatistics, Graduate School of Medicine, Kyoto University
| | - Morio Sawamura
- Department of Hematology, National Hospital Organization Shibukawa Medical Center
| | - Michihiro Hidaka
- Department of Hematology, National Hospital Organization Kumamoto Medical Center
| | - Shinichiro Yoshida
- Department of Hematology, National Hospital Organization Nagasaki Medical Center
| | - Naokuni Uike
- Department of Hematology, National Hospital Organization Kyushu Cancer Center
| | - Yukihiro Kaneko
- Department of Bacteriology, Osaka City University Graduate School of Medicine.,Research Center for Infectious Disease Sciences, Osaka City University Graduate School of Medicine.,Department of Chemotherapy and Mycoses, National Institute of Infectious Diseases
| | - Yoshitsugu Miyazaki
- Department of Chemotherapy and Mycoses, National Institute of Infectious Diseases
| | - Hirokazu Nagai
- Department of Hematology, National Hospital Organization Nagoya Medical Center
| |
Collapse
|
30
|
Tragiannidis A, Gkampeta A, Vousvouki M, Vasileiou E, Groll AH. Antifungal agents and the kidney: pharmacokinetics, clinical nephrotoxicity, and interactions. Expert Opin Drug Saf 2021; 20:1061-1074. [PMID: 33896310 DOI: 10.1080/14740338.2021.1922667] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
INTRODUCTION Invasive fungal infections continue to be important causes of morbidity and mortality in severely ill and immunocompromised patient populations. The past three decades have seen a considerable expansion in antifungal drug research, resulting in the clinical development of different classes of antifungal agents with different pharmacologic properties. Among drug-specific characteristics of antifungal agents, renal disposition and nephrotoxicity are important clinical considerations as many patients requiring antifungal therapy have compromised organ functions or are receiving other potentially nephrotoxic medications. AREAS COVERED The present article reviews incidence, severity and mechanisms of nephrotoxicity associated with antifungal agents used for prevention and treatment of invasive fungal diseases by discussing distribution, metabolism, elimination and drug-related adverse events in the context of safety data from phase II and III clinical studies. EXPERT OPINION Based on the available data amphotericin B deoxycholate has the highest relative potential for nephrotoxicity, followed by the lipid formulations of amphotericin B, and, to a much lesser extent and by indirect mechanisms, the antifungal triazoles.
Collapse
Affiliation(s)
- Athanasios Tragiannidis
- Childhood & Adolescent Hematology Oncology Unit, 2nd Pediatric Department, Faculty of Health Sciences, Aristotle University of Thessaloniki, AHEPA Hospital, Greece
- Infectious Disease Research Program, Center for Bone Marrow Transplantation and Department of Pediatric Hematology/Oncology, University Children's Hospital Münster, Münster, Germany
| | - Anastasia Gkampeta
- Childhood & Adolescent Hematology Oncology Unit, 2nd Pediatric Department, Faculty of Health Sciences, Aristotle University of Thessaloniki, AHEPA Hospital, Greece
| | - Maria Vousvouki
- Childhood & Adolescent Hematology Oncology Unit, 2nd Pediatric Department, Faculty of Health Sciences, Aristotle University of Thessaloniki, AHEPA Hospital, Greece
| | - Eleni Vasileiou
- Childhood & Adolescent Hematology Oncology Unit, 2nd Pediatric Department, Faculty of Health Sciences, Aristotle University of Thessaloniki, AHEPA Hospital, Greece
| | - Andreas H Groll
- Infectious Disease Research Program, Center for Bone Marrow Transplantation and Department of Pediatric Hematology/Oncology, University Children's Hospital Münster, Münster, Germany
| |
Collapse
|
31
|
Rahi MS, Jindal V, Pednekar P, Parekh J, Gunasekaran K, Sharma S, Stender M, Jaiyesimi IA. Fungal infections in hematopoietic stem-cell transplant patients: a review of epidemiology, diagnosis, and management. Ther Adv Infect Dis 2021; 8:20499361211039050. [PMID: 34434551 PMCID: PMC8381463 DOI: 10.1177/20499361211039050] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 07/24/2021] [Indexed: 12/18/2022] Open
Abstract
The advent of bone marrow transplant has opened doors to a different approach and
offered a new treatment modality for various hematopoietic stem-cell-related
disorders. Since the first bone marrow transplant in 1957, there has been
significant progress in managing patients who undergo bone marrow transplants.
Plasma-cell disorders, lymphoproliferative disorders, and myelodysplastic
syndrome are the most common indications for hematopoietic stem-cell transplant.
Despite the advances, invasive fungal infections remain a significant cause of
morbidity and mortality in this high-risk population. The overall incidence of
invasive fungal infection in patients with hematopoietic stem-cell transplant is
around 4%, but the mortality in patients with allogeneic stem-cell transplant is
as high as 13% in one study. Type of stem-cell transplant, conditioning regimen,
and development of graft-versus-host disease are some of the
risk factors that impact the risk and outcomes in patients with invasive fungal
infections. Aspergillus and candida remain the two most common organisms causing
invasive fungal infections. Molecular diagnostic methods have replaced some
traditional methods due to their simplicity of use and rapid turnaround time.
Primary prophylaxis has undoubtedly shown to improve outcomes even though
breakthrough infection rates remain high. The directed treatment has seen a
significant shift from amphotericin B to itraconazole, voriconazole, and
echinocandins, which have shown better efficacy and fewer adverse effects. In
this comprehensive review, we aim to detail epidemiology, risk factors,
diagnosis, and management, including prophylaxis, empiric and directed
management of invasive fungal infections in patients with hematopoietic
stem-cell transplant.
Collapse
Affiliation(s)
- Mandeep Singh Rahi
- Division of Pulmonary Diseases and Critical Care Medicine, Yale-New Haven Health Bridgeport Hospital, 267 Grant Street, Bridgeport, CT 06610, USA
| | - Vishal Jindal
- Division of Hematology and Oncology, Oakland University-William Beaumont School of Medicine, Royal Oak, MI, USA
| | - Prachi Pednekar
- Department of Internal Medicine, Yale-New Haven Health Bridgeport Hospital, Bridgeport, CT, USA
| | - Jay Parekh
- Department of Internal Medicine, Yale-New Haven Health Bridgeport Hospital, Bridgeport, CT, USA
| | - Kulothungan Gunasekaran
- Division of Pulmonary Diseases and Critical Care Medicine, Yale-New Haven Health Bridgeport Hospital, Bridgeport, CT, USA
| | - Sorabh Sharma
- Department of Internal Medicine, Banner University Medical Center, Tucson, AZ, USA
| | - Michael Stender
- Division of Hematology and Oncology, Oakland University-William Beaumont School of Medicine, Royal Oak, MI, USA
| | - Ishmael A Jaiyesimi
- Division of Hematology and Oncology, Oakland University-William Beaumont School of Medicine, Royal Oak, MI, USA
| |
Collapse
|
32
|
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
|
33
|
Kato H, Hagihara M, Shibata Y, Asai N, Yamagishi Y, Iwamoto T, Mikamo H. Comparison of mortality between echinocandins and polyenes for an initial treatment of candidemia: A systematic review and meta-analysis. J Infect Chemother 2021; 27:1562-1570. [PMID: 34217605 DOI: 10.1016/j.jiac.2021.06.017] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 06/14/2021] [Accepted: 06/21/2021] [Indexed: 11/28/2022]
Abstract
BACKGROUND Current guidelines recommend echinocandins for the initial treatment of candidemia. However, polyenes are often chosen in clinical settings because of their fungicidal and anti-biofilm effects. Therefore, we performed a systematic review and meta-analysis to evaluate whether echinocandins are superior to polyenes in terms of mortality for the initial treatment of candidemia. METHODS We systematically searched the Scopus, EMBASE, Cochrane Central Register of Controlled Trials, PubMed, and CINAHL databases until July 1, 2020. We compared the mortality rates of patients who received echinocandins and polyenes. As a subgroup analysis, we compared the mortality rates following the use of echinocandins versus liposomal amphotericin B. RESULTS Fifteen studies involving 854 patients were included. Various Candida species were detected, and the rates of resistance of echinocandins and polyenes against the overall detected isolates were 1.0% and 0%, respectively. The overall mortality recorded in 15 studies was 41.0%, and the mortality was significantly higher for polyenes than echinocandins (odd ratios [OR] 1.68, 95% confidential interval [CI] 1.17-2.42). Furthermore, liposomal amphotericin B showed higher mortality in the initial treatment than echinocandins (OR 1.42; 95% CI 0.84-2.39). CONCLUSIONS We revealed an association between echinocandin treatment and reduced mortality in the initial treatment of candidemia when causative fungi were not considered. Our findings partially support current guidelines recommending echinocandins for the treatment of candidemia.
Collapse
Affiliation(s)
- Hideo Kato
- Department of Clinical Infectious Diseases, Aichi Medical University, Aichi, Japan; Department of Pharmacy, Mie University Hospital, Mie, Japan
| | - Mao Hagihara
- Department of Clinical Infectious Diseases, Aichi Medical University, Aichi, Japan; Department of Molecular Epidemiology and Biomedical Sciences, Aichi Medical University Hospital, Aichi, Japan
| | - Yuichi Shibata
- Department of Clinical Infectious Diseases, Aichi Medical University, Aichi, Japan
| | - Nobuhiro Asai
- Department of Clinical Infectious Diseases, Aichi Medical University, Aichi, Japan
| | - Yuka Yamagishi
- Department of Clinical Infectious Diseases, Aichi Medical University, Aichi, Japan
| | - Takuya Iwamoto
- Department of Pharmacy, Mie University Hospital, Mie, Japan
| | - Hiroshige Mikamo
- Department of Clinical Infectious Diseases, Aichi Medical University, Aichi, Japan.
| |
Collapse
|
34
|
Abstract
Over the past 15 years, there has been an increase in the development and utilization of newer antifungal agents. The ideal antifungal, however, in regard to spectrum of activity, pharmacokinetic/pharmacodynamic properties, development of resistance, safety, and drug interaction profile remains elusive. This article reviews pharmacologic aspects of Food and Drug Administration-approved polyenes, flucytosine, azoles, and echinocandins as well as promising pipeline antifungal agents. Unique properties of these newer agents are highlighted. The clinical role of established and investigational antifungal agents as treatment and/or prevention of invasive fungal infections is discussed.
Collapse
Affiliation(s)
- Melissa D Johnson
- Duke University Medical Center, Box 102359 DUMC, Durham NC 27710, USA.
| |
Collapse
|
35
|
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: 0.5] [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
|
36
|
Kobayashi R, Matsushima S, Hori D, Sano H, Suzuki D, Kishimoto K, Nakano T, Yanagi M, Kodama K, Kobayashi K. Efficacy of liposomal amphotericin against febrile neutropenia in pediatric patients receiving prophylactic voriconazole. Pediatr Int 2021; 63:550-555. [PMID: 32869416 DOI: 10.1111/ped.14450] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 08/13/2020] [Accepted: 08/20/2020] [Indexed: 11/28/2022]
Abstract
BACKGROUND The risk factors for invasive fungal infection have gradually become evident for pediatric patients with hematological diseases. Here we analyze the efficacy of liposomal amphotericin (L-AMB) for pediatric patients with febrile neutropenia using prophylactic voriconazole (VRCZ). METHOD We administered L-AMB (2.5 mg/kg/day) in patients with febrile neutropenia who were receiving prophylactic VRCZ (10 mg/kg/day, orally) and were resistant to second-line antibiotics therapy. Thirteen patients (5 males, 8 females) with 19 febrile neutropenia episodes were targeted in this analysis. The median age of the patients was 14 years (range, 1-19 years). Eighteen out of 19 episodes occurred in patients with acute myeloid leukemia, with the remaining episode occurring in a patient with acute unclassified leukemia. RESULTS The median period from start of L-AMB administration to resolution of fever was 4 days (1-27 days). In 15 out of 19 episodes, fever resolved within 5 days from commencement of L-AMB administration. Using criteria proposed by T. J. Walsh et al., the success rate of L-AMB for febrile neutropenia was 89.5% in this study. CONCLUSIONS Although the sample size of our study was small, the extremely high efficacy of L-AMB warrants its administration in patients with febrile neutropenia who are receiving VRCZ.
Collapse
Affiliation(s)
- Ryoji Kobayashi
- Department of Hematology/Oncology for Children and Adolescents, Sapporo Hokuyu Hospital, Sapporo, Japan
| | - Satoru Matsushima
- Department of Hematology/Oncology for Children and Adolescents, Sapporo Hokuyu Hospital, Sapporo, Japan
| | - Daiki Hori
- Department of Hematology/Oncology for Children and Adolescents, Sapporo Hokuyu Hospital, Sapporo, Japan
| | - Hirozumi Sano
- Department of Hematology/Oncology for Children and Adolescents, Sapporo Hokuyu Hospital, Sapporo, Japan
| | - Daisuke Suzuki
- Department of Hematology/Oncology for Children and Adolescents, Sapporo Hokuyu Hospital, Sapporo, Japan
| | - Kenji Kishimoto
- Department of Hematology/Oncology for Children and Adolescents, Sapporo Hokuyu Hospital, Sapporo, Japan
| | - Takaaki Nakano
- Department of Hematology/Oncology for Children and Adolescents, Sapporo Hokuyu Hospital, Sapporo, Japan
| | - Masato Yanagi
- Department of Hematology/Oncology for Children and Adolescents, Sapporo Hokuyu Hospital, Sapporo, Japan
| | - Kouya Kodama
- Department of Hematology/Oncology for Children and Adolescents, Sapporo Hokuyu Hospital, Sapporo, Japan
| | - Kunihiko Kobayashi
- Department of Hematology/Oncology for Children and Adolescents, Sapporo Hokuyu Hospital, Sapporo, Japan
| |
Collapse
|
37
|
8th European Conference on Infections in Leukaemia: 2020 guidelines for the diagnosis, prevention, and treatment of invasive fungal diseases in paediatric patients with cancer or post-haematopoietic cell transplantation. Lancet Oncol 2021; 22:e254-e269. [PMID: 33811813 DOI: 10.1016/s1470-2045(20)30723-3] [Citation(s) in RCA: 126] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 11/16/2020] [Accepted: 11/17/2020] [Indexed: 11/24/2022]
Abstract
Paediatric patients with cancer and those undergoing allogeneic haematopoietic cell transplantation have an increased susceptibility to invasive fungal diseases. In addition to differences in underlying conditions and comorbidities relative to adults, invasive fungal diseases in infants, children, and adolescents are unique in terms of their epidemiology, the validity of current diagnostic methods, the pharmacology and dosing of antifungal agents, and the absence of phase 3 clinical trials to provide data to guide evidence-based interventions. To re-examine the state of knowledge and to further improve invasive fungal disease diagnosis, prevention, and management, the 8th European Conference on Infections in Leukaemia (ECIL-8) reconvened a Paediatric Group to review the literature and to formulate updated recommendations according to the European Society of Clinical Microbiology and Infectious Diseases (ESCMID) and European Confederation of Medical Mycology (ECMM) grading system, which are summarised in this Review.
Collapse
|
38
|
[Consensus of Chinese experts on prevention and standardized treatment of drug-induced liver injury in patients with blood diseases (2021)]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2021; 42:185-192. [PMID: 33910302 PMCID: PMC8081943 DOI: 10.3760/cma.j.issn.0253-2727.2021.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Indexed: 11/05/2022]
|
39
|
Abstract
PURPOSE OF REVIEW To highlight recent findings on the adequate duration of antifungal therapy in patients with invasive fungal disease (IFD). RECENT FINDINGS Plenty of published data available suggest that there is no additional clinical benefit at a certain point after initiation of antifungal treatment in patients with confirmed IFD. Moreover, the prolonged antifungal exposure can be associated with an increased risk of side effects and toxicity as well as striking risk for developing antifungal resistance or rising unnecessary healthcare costs. Recent data suggest that, in the presence of an adequate initial antifungal therapy and adequate source control of the infection, new stratified approaches integrating clinical judgment, biomarkers and microbiological eradication, should be considered as an alternative to the 'one-size-fits-all' treatment duration currently used worldwide. SUMMARY The optimal duration of antifungal therapy is still an unresolved issue that depends by many key elements including the host; the pathogen and its microbiological eradication, the adequateness of initial antifungal therapy and the promptness of source control of the infection. In general, many patients with invasive candidiasis can be treated with a 2 weeks course of antifungal therapy. Longer antifungal course (6 weeks or more) is generally required for patients with invasive aspergilosis.
Collapse
|
40
|
Zheng XY, Liang AB, Yang XZ, Fu JF, Hou M, Sun AN, Lu H, Jin J, Hu JD. [Pharmacokinetic study of domestic caspofungin compared with original caspofungin for empirical therapy in patients with persistent fever and agranulocytosis]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2020; 41:1031-1034. [PMID: 33445852 PMCID: PMC7840557 DOI: 10.3760/cma.j.issn.0253-2727.2020.12.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Indexed: 11/05/2022]
Affiliation(s)
- X Y Zheng
- Department of Hematology, Fujian Medical University Union Hospital, Fujian Institute of Hematology, Fujian Provincial Key Laboratory of Hematology, Fuzhou 350001, China
| | - A B Liang
- Department of Hematology, Tongji Hospital, Tongji University, Shanghai 200065, China
| | - X Z Yang
- Department of Hematology, Fujian Medical University Union Hospital, Fujian Institute of Hematology, Fujian Provincial Key Laboratory of Hematology, Fuzhou 350001, China
| | - J F Fu
- Department of Hematology, Tongji Hospital, Tongji University, Shanghai 200065, China
| | - M Hou
- Department of Hematology, Qilu Hospital, Shandong University, Jinan 250012, China
| | - A N Sun
- Department of Hematology, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - H Lu
- Department of Hematology, Jiangsu Province Hospital, Nanjing 210029, China
| | - J Jin
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - J D Hu
- Department of Hematology, Fujian Medical University Union Hospital, Fujian Institute of Hematology, Fujian Provincial Key Laboratory of Hematology, Fuzhou 350001, China
| |
Collapse
|
41
|
A real-world prospective observational study on the efficacy and safety of liposomal amphotericin B in 426 patients with persistent neutropenia and fever. J Infect Chemother 2020; 27:277-283. [PMID: 33109439 DOI: 10.1016/j.jiac.2020.10.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Revised: 10/04/2020] [Accepted: 10/06/2020] [Indexed: 11/20/2022]
Abstract
INTRODUCTION Invasive fungal diseases are crucial causes of morbidity and mortality among patients with febrile neutropenia (FN). Though liposomal amphotericin B (L-AMB) is one of the agents recommended for first-line empirical antifungal therapy in patients with FN, large-scale clinical studies have not been performed in Japan. METHODS An open-label prospective multi-center study was carried out to evaluate the safety and efficacy of L-AMB in Japanese patients with FN suspected of having fungal infection. RESULTS Of the 426 patients registered, safety and efficacy evaluations were conducted for 424 and 399, respectively. By clinical response criteria using 5 composite endpoints, the response rate was 46.6% (186/399). The response rate by age were 54.5% (child: 30/55), 47.5% (adult: 97/204), 42.1% (elderly: 59/140) respectively. Regarding the composite endpoints, resolution of fever was observed in 61.2% (244/399), no breakthrough fungal infection in 99.0% (395/399), survival for 7 days or longer after the completion of treatment in 83.7% (334/399), no discontinuation of treatment due to toxicity or lack of efficacy in 60.9% (243/399), and successful treatment of any baseline fungal infection in 10/18. Adverse drug reactions (ADRs) developed in 61.1% (259/424), and frequent ADRs were hypokalemia, kidney dysfunction, and liver dysfunction, as previously reported. CONCLUSIONS The safety and efficacy profile of L-AMB in Japanese patients with FN suspected of having fungal infection were elucidated for the first time, through the analysis of a large number of cases including pediatric patients under real-world clinical settings collected in this nationwide study.
Collapse
|
42
|
Christopeit M, Schmidt-Hieber M, Sprute R, Buchheidt D, Hentrich M, Karthaus M, Penack O, Ruhnke M, Weissinger F, Cornely OA, Maschmeyer G. Prophylaxis, diagnosis and therapy of infections in patients undergoing high-dose chemotherapy and autologous haematopoietic stem cell transplantation. 2020 update of the recommendations of the Infectious Diseases Working Party (AGIHO) of the German Society of Hematology and Medical Oncology (DGHO). Ann Hematol 2020; 100:321-336. [PMID: 33079221 PMCID: PMC7572248 DOI: 10.1007/s00277-020-04297-8] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 10/03/2020] [Indexed: 12/14/2022]
Abstract
To ensure the safety of high-dose chemotherapy and autologous stem cell transplantation (HDC/ASCT), evidence-based recommendations on infectious complications after HDC/ASCT are given. This guideline not only focuses on patients with haematological malignancies but also addresses the specifics of HDC/ASCT patients with solid tumours or autoimmune disorders. In addition to HBV and HCV, HEV screening is nowadays mandatory prior to ASCT. For patients with HBs antigen and/or anti-HBc antibody positivity, HBV nucleic acid testing is strongly recommended for 6 months after HDC/ASCT or for the duration of a respective maintenance therapy. Prevention of VZV reactivation by vaccination is strongly recommended. Cotrimoxazole for the prevention of Pneumocystis jirovecii is supported. Invasive fungal diseases are less frequent after HDC/ASCT, therefore, primary systemic antifungal prophylaxis is not recommended. Data do not support a benefit of protective room ventilation e.g. HEPA filtration. Thus, AGIHO only supports this technique with marginal strength. Fluoroquinolone prophylaxis is recommended to prevent bacterial infections, although a survival advantage has not been demonstrated.
Collapse
Affiliation(s)
- Maximilian Christopeit
- Department of Stem Cell Transplantation, University Medical Center Eppendorf, Hamburg, Germany.
| | - Martin Schmidt-Hieber
- Department of Hematology and Oncology, Carl-Thiem-Klinikum, Cottbus, Cottbus, Germany
| | - Rosanne Sprute
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
- Department I of Internal Medicine, University Hospital of Cologne, University of Cologne, Cologne, Germany
- Partner Site Bonn-Cologne, German Centre for Infection Research, Cologne, Germany
| | - Dieter Buchheidt
- Department of Hematology and Oncology, Mannheim University Hospital, Heidelberg University, Mannheim, Germany
| | - Marcus Hentrich
- Department of Medicine III-Hematology/Oncology, Red Cross Hospital, Munich, Germany
| | - Meinolf Karthaus
- Department of Internal Medicine, Hematology and Oncology, Klinikum Neuperlach, Städtisches Klinikum München, Munich, Germany
| | - Olaf Penack
- Department of Internal Medicine, Division of Hematology and Oncology, Charité Universitätsmedizin Berlin, Campus Rudolf Virchow, Berlin, Germany
| | - Markus Ruhnke
- Department of Hematology, Oncology and Palliative Medicine, Helios Hospital Aue, Aue, Germany
| | - Florian Weissinger
- Department of Internal Medicine, Hematology, Oncology, Stem Cell Transplantation and Palliative Medicine, Protestant Hospital of Bethel Foundation, Bielefeld, Germany
| | - 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, University Hospital of Cologne, University of Cologne, Cologne, Germany
- Partner Site Bonn-Cologne, German Centre for Infection Research, Cologne, Germany
- Clinical Trials Centre Cologne (ZKS Köln), University of Cologne, Cologne, Germany
| | - Georg Maschmeyer
- Klinikum Ernst von Bergmann, Department of Hematology, Oncology and Palliative Care, Potsdam, Germany
| |
Collapse
|
43
|
Peseski AM, McClean M, Green SD, Beeler C, Konig H. Management of fever and neutropenia in the adult patient with acute myeloid leukemia. Expert Rev Anti Infect Ther 2020; 19:359-378. [PMID: 32892669 DOI: 10.1080/14787210.2020.1820863] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
INTRODUCTION Febrile neutropenia represents one of the most common treatment-associated complications in the management of acute myeloid leukemia (AML) and is considered an oncologic emergency. Rapid and detailed workup as well as the initiation of empiric broad-spectrum antibiotic therapy are critical to avoid sepsis and to reduce mortality. Although a definitive source of infection is frequently not identified, the severely immunosuppressed status of the AML patient undergoing cytotoxic therapy results in a high risk for a wide array of bacterial, fungal, and viral etiologies. AREAS COVERED The authors herein review the diagnostic and therapeutic approach to the neutropenic leukemia patient based on the current knowledge. Special consideration is given to the rapidly changing therapeutic landscape in AML, creating new challenges in the management of infectious complications. EXPERT OPINION Multidrug-resistant organisms pose a major challenge in the management of neutropenic fever patients with hematologic malignancies - including AML. Future directions to improve outcomes demand innovative treatment approaches as well as advances in biomarker research to facilitate diagnosis and disease monitoring. Recent achievements in AML-targeted therapy led to an increased incidence of differentiation syndrome, a potentially life-threatening side effect that frequently resembles clinical infection and requires prompt recognition and aggressive intervention.
Collapse
Affiliation(s)
- Andrew M Peseski
- Department of Internal Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Mitchell McClean
- Department of Internal Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Steven D Green
- Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Cole Beeler
- Division of Infectious Diseases, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Heiko Konig
- Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
44
|
Modeling Invasive Aspergillosis: How Close Are Predicted Antifungal Targets? J Fungi (Basel) 2020; 6:jof6040198. [PMID: 33007839 PMCID: PMC7712059 DOI: 10.3390/jof6040198] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 09/23/2020] [Accepted: 09/25/2020] [Indexed: 02/06/2023] Open
Abstract
Animal model systems are a critical component of the process of discovery and development of new antifungal agents for treatment and prevention of invasive aspergillosis. The persistently neutropenic rabbit model of invasive pulmonary aspergillosis (IPA) has been a highly predictive system in identifying new antifungal agents for treatment and prevention of this frequently lethal infection. Since its initial development, the persistently neutropenic rabbit model of IPA has established a strong preclinical foundation for dosages, drug disposition, pharmacokinetics, safety, tolerability, and efficacy for deoxycholate amphotericin B, liposomal amphotericin B, amphotericin B lipid complex, amphotericin B colloidal dispersion, caspofungin, micafungin, anidulafungin, voriconazole, posaconazole, isavuconazole, and ibrexafungerp in treatment of patients with invasive aspergillosis. The findings of combination therapy with a mould-active triazole and an echinocandin in this rabbit model also predicted the outcome of the clinical trial for voriconazole plus anidulafungin for treatment of IPA. The plasma pharmacokinetic parameters and tissue disposition for most antifungal agents approximate those of humans in persistently neutropenic rabbits. Safety, particularly nephrotoxicity, has also been highly predictive in the rabbit model, as exemplified by the differential glomerular filtration rates observed in animals treated with deoxycholate amphotericin B, liposomal amphotericin B, amphotericin B lipid complex, and amphotericin B colloidal dispersion. A panel of validated outcome variables measures therapeutic outcome in the rabbit model: residual fungal burden, markers of organism-mediated pulmonary injury (lung weights and infarct scores), survival, and serum biomarkers. In selected antifungal studies, thoracic computerized tomography (CT) is also used with diagnostic imaging algorithms to measure therapeutic response of pulmonary infiltrates, which exhibit characteristic radiographic patterns, including nodules and halo signs. Further strengthening the predictive properties of the model, therapeutic response to successfully developed antifungal agents for treatment of IPA has been demonstrated over the past two decades by biomarkers of serum galactomannan and (1→3)-β-D-glucan with patterns of resolution, that closely mirror those documented responses in patients with IPA. The decision to move from laboratory to clinical trials should be predicated upon a portfolio of complementary and mutually validating preclinical laboratory animal models studies. Other model systems, including those in mice, rats, and guinea pigs, are also valuable tools in developing clinical protocols. Meticulous preclinical investigation of a candidate antifungal compound in a robust series of complementary laboratory animal models will optimize study design, de-risk clinical trials, and ensure tangible benefit to our most vulnerable immunocompromised patients with invasive aspergillosis.
Collapse
|
45
|
Salmanton-García J, Seidel D, Koehler P, Mellinghoff SC, Herbrecht R, Klimko N, Ráčil Z, Falces-Romero I, Ingram P, Benítez-Peñuela MÁ, Rodríguez JY, Desoubeaux G, Barać A, García-Vidal C, Hoenigl M, Mehta SR, Cheng MP, Klyasova G, Heinz WJ, Iqbal N, Krause R, Ostermann H, Penack O, Schalk E, Sheppard DC, Willinger B, Wisplinghoff H, Vehreschild JJ, Cornely OA, Vehreschild MJGT. Matched-paired analysis of patients treated for invasive mucormycosis: standard treatment versus posaconazole new formulations (MoveOn). J Antimicrob Chemother 2020; 74:3315-3327. [PMID: 31393591 DOI: 10.1093/jac/dkz344] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Revised: 07/04/2019] [Accepted: 07/09/2019] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND First-line antifungal treatment for invasive mucormycosis (IM) consists of liposomal amphotericin B. Salvage treatment options are limited and often based on posaconazole oral suspension. With the approval of posaconazole new formulations, patients could benefit from improved pharmacokinetics, safety and tolerability. OBJECTIVES Our aim was to assess the effectiveness of posaconazole new formulations for IM treatment. METHODS We performed a case-matched analysis with proven or probable IM patients from the FungiScope® Registry. First-line posaconazole new formulations (1st-POSnew) and first-line amphotericin B plus posaconazole new formulations (1st-AMB+POSnew) cases were matched with first-line amphotericin B-based (1st-AMB) treatment controls. Salvage posaconazole new formulations (SAL-POSnew) cases were matched with salvage posaconazole oral suspension (SAL-POSsusp) controls. Each case was matched with up to three controls (based on severity, haematological/oncological malignancy, surgery and/or renal dysfunction). RESULTS Five patients receiving 1st-POSnew, 18 receiving 1st-AMB+POSnew and 22 receiving SAL-POSnew were identified. By day 42, a favourable response was reported for 80.0% (n = 4/5) of patients receiving 1st-POSnew, for 27.8% (n = 5/18) receiving 1st-AMB+POSnew and for 50.0% (n = 11/22) receiving SAL-POSnew. Day 42 all-cause mortality of patients receiving posaconazole new formulations was lower compared with controls [20.0% (n = 1/5) in 1st-POSnew versus 53.3% (n = 8/15) in 1st-AMB; 33.3% (n = 6/18) in 1st-AMB+POSnew versus 52.0% (n = 26/50) in 1st-AMB; and 0.0% (n = 0/22) in SAL-POSnew versus 4.4% (n = 2/45) in SAL-POSsusp]. CONCLUSIONS Posaconazole new formulations were effective in terms of treatment response and associated mortality of IM. While posaconazole new formulations may be an alternative for treatment of IM, the limited sample size of our study calls for a cautious interpretation of these observations.
Collapse
Affiliation(s)
- Jon Salmanton-García
- University of Cologne, Faculty of Medicine and University Hospital of Cologne, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD), European Diamond Excellence Center for Medical Mycology (ECMM), Cologne, Germany
| | - Danila Seidel
- University of Cologne, Faculty of Medicine and University Hospital of Cologne, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD), European Diamond Excellence Center for Medical Mycology (ECMM), Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Philipp Koehler
- University of Cologne, Faculty of Medicine and University Hospital of Cologne, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD), European Diamond Excellence Center for Medical Mycology (ECMM), Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Sibylle C Mellinghoff
- University of Cologne, Faculty of Medicine and University Hospital of Cologne, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD), European Diamond Excellence Center for Medical Mycology (ECMM), Cologne, Germany
| | - Raoul Herbrecht
- Department of Oncology and Hematology, Hôpitaux Universitaires de Strasbourg and Université de Strasbourg, Inserm, UMR-S1113/IRFAC, Strasbourg, France
| | - Nikolai Klimko
- Department of Clinical Mycology, Allergy and Immunology, North Western State Medical University, Saint Petersburg, Russia
| | - Zdeněk Ráčil
- Department of Internal Medicine-Hematology and Oncology, Masaryk University, Brno, Czech Republic.,University Hospital Brno, Brno, Czech Republic
| | - Iker Falces-Romero
- Clinical Microbiology and Parasitology Department, University Hospital La Paz, Madrid, Spain
| | - Paul Ingram
- Department of Infectious Diseases, Royal Perth Hospital, Perth, WA, Australia.,School of Pathology and Laboratory Medicine, University of Western Australia, Perth, WA, Australia
| | - Miguel-Ángel Benítez-Peñuela
- Center of Microbiological Research of Cesar (CIMCE), Rosario Pumarejo de López Hospital, Laura Daniela Clinic, Médicos Clinic LTDA, Valledupar, Colombia
| | - José Yesid Rodríguez
- Center of Microbiological Research of Cesar (CIMCE), Rosario Pumarejo de López Hospital, Laura Daniela Clinic, Médicos Clinic LTDA, Valledupar, Colombia
| | - Guillaume Desoubeaux
- Parasitology, Mycology and Tropical Medicine Service, University Hospital of Tours, Tours, France.,Inserm U1100, Tours University, Tours, France
| | - Aleksandra Barać
- Clinic for Infectious and Tropical Diseases, Clinical Centre of Serbia, Faculty of Medicine, University of Belgrade, Serbia
| | - Carolina García-Vidal
- Service of Infectious Diseases, Clínic Hospital, University of Barcelona, Institute of Biomedical Research August Pi i Sunyer, Barcelona, Spain
| | - Martin Hoenigl
- Division of Infectious Diseases, University of California San Diego, San Diego, CA, USA.,Section of Infectious Diseases and Tropical Medicine, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Sanjay R Mehta
- Division of Infectious Diseases, University of California San Diego, San Diego, CA, USA.,Department of Medicine, San Diego Veterans Affairs Medical Center San Diego, CA, USA
| | - Matthew P Cheng
- Division of Infectious Diseases, Departments of Medicine, Microbiology and Immunology, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Galina Klyasova
- Department of Clinical Microbiology, Mycology and Antibiotic Therapy, National Research Center for Hematology, Moscow, Russia
| | - Werner J Heinz
- Department of Internal Medicine II, Julius Maximilians University, Würzburg, Germany
| | - Nousheen Iqbal
- Section of Pulmonology and Critical Care, Department of Medicine, Aga Khan University Hospital, Karachi, Pakistan
| | - Robert Krause
- Section of Infectious Diseases and Tropical Medicine, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Helmut Ostermann
- Department of Internal Medicine III, University of Munich, Munich, Germany
| | - Olaf Penack
- Department for Hematology, Oncology and Tumorimmunology, Charité University Medicine Berlin, Campus Virchow Clinic, Berlin, Germany
| | - Enrico Schalk
- Department of Haematology and Oncology, Medical Centre, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Donald C Sheppard
- Division of Infectious Diseases, Departments of Medicine, Microbiology and Immunology, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Birgit Willinger
- Division of Clinical Microbiology, Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Hilmar Wisplinghoff
- University of Cologne, Institute for Medical Microbiology, Immunology and Hygiene, Cologne, Germany.,Wisplinghoff Laboratories, Cologne, Germany.,Institute for Virology and Clinical Microbiology, Witten/Herdecke University, Witten, Germany
| | - J Janne Vehreschild
- University of Cologne, Faculty of Medicine and University Hospital of Cologne, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD), European Diamond Excellence Center for Medical Mycology (ECMM), Cologne, Germany.,German Centre for Infection Research (DZIF), partner site Bonn - Cologne, Cologne, Germany.,Center for Integrated Oncology CIO Köln/Bonn, Medical Faculty, University of Cologne, Cologne, Germany
| | - Oliver A Cornely
- University of Cologne, Faculty of Medicine and University Hospital of Cologne, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD), European Diamond Excellence Center for Medical Mycology (ECMM), Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany.,German Centre for Infection Research (DZIF), partner site Bonn - Cologne, Cologne, Germany.,Center for Integrated Oncology CIO Köln/Bonn, Medical Faculty, University of Cologne, Cologne, Germany.,Clinical Trials Centre Cologne (ZKS Köln), University of Cologne, Cologne, Germany.,Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Maria J G T Vehreschild
- University of Cologne, Faculty of Medicine and University Hospital of Cologne, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD), European Diamond Excellence Center for Medical Mycology (ECMM), Cologne, Germany.,German Centre for Infection Research (DZIF), partner site Bonn - Cologne, Cologne, Germany.,Center for Integrated Oncology CIO Köln/Bonn, Medical Faculty, University of Cologne, Cologne, Germany.,Department of Internal Medicine, Infectious Diseases, Goethe University, Frankfurt, Frankfurt am Main, Germany
| | | |
Collapse
|
46
|
Kimura SI, Kanda Y, Iino M, Fukuda T, Sakaida E, Oyake T, Yamaguchi H, Fujiwara SI, Jo Y, Okamoto A, Fujita H, Takamatsu Y, Saburi Y, Matsumura I, Yamanouchi J, Shiratori S, Gotoh M, Nakamura S, Tamura K. Efficacy and safety of micafungin in empiric and D-index-guided early antifungal therapy for febrile neutropenia; A subgroup analysis of the CEDMIC trial. Int J Infect Dis 2020; 100:292-297. [PMID: 32891738 DOI: 10.1016/j.ijid.2020.08.081] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 08/23/2020] [Accepted: 08/25/2020] [Indexed: 10/23/2022] Open
Abstract
OBJECTIVES The D-index is defined as the area over the neutrophil curve during neutropenia. The CEDMIC trial confirmed the noninferiority of D-index-guided early antifungal therapy (DET) using micafungin to empirical antifungal therapy (EAT). In this study, we evaluated the efficacy and safety of micafungin in these settings. METHODS From the CEDMIC trial, we extracted 67 and 113 patients who received micafungin in the DET and EAT groups, respectively. Treatment success was defined as the fulfilment of all components of a five-part composite end point. Fever resolution was evaluated at seven days after the completion of therapy. RESULTS The proportion of high-risk treatments including induction chemotherapy for acute leukemia and allogeneic hematopoietic stem cell transplantation was significantly higher in the DET group than in the EAT group (82.1% vs. 52.2%). The efficacy of micafungin was 68.7% (95%CI: 56.2-79.4) and 79.6% (71.0-86.6) in the DET and EAT groups, respectively. When we focused on high-risk treatments, the efficacy was 69.1% (55.2-80.9%) and 78.0% (65.3-87.7%), respectively (P = 0.30). There was no significant difference in any of the 5 components between the two groups. CONCLUSIONS The efficacy of micafungin in patients undergoing high-risk treatment was not strongly impaired in DET compared to that in EAT.
Collapse
Affiliation(s)
- Shun-Ichi Kimura
- Division of Hematology, Jichi Medical University Saitama Medical Center, Japan
| | - Yoshinobu Kanda
- Division of Hematology, Jichi Medical University Saitama Medical Center, Japan; Division of Hematology, Department of Medicine, Jichi Medical University, Japan.
| | - Masaki Iino
- Department of Medical Oncology, Yamanashi Prefectural Central Hospital, Japan
| | - Takahiro Fukuda
- Department of Hematopoietic Stem Cell Transplantation, National Cancer Center Hospital, Japan
| | - Emiko Sakaida
- Department of Hematology, Chiba University Hospital, Japan
| | - Tatsuo Oyake
- Division of Hematology and Oncology, Department of Internal Medicine, Iwate Medical University School of Medicine, Japan
| | | | | | - Yumi Jo
- Department of Oncology and Hematology / Infection Control Division, Shimane University Hospital, Japan
| | - Akinao Okamoto
- Department of Hematology, Fujita Health University School of Medicine, Japan
| | - Hiroyuki Fujita
- Department of Hematology, Saiseikai Yokohama Nanbu Hospital, Japan
| | - Yasushi Takamatsu
- Division of Medical Oncology, Hematology and Infectious Diseases, Department of Internal Medicine, Fukuoka University Hospital, Japan
| | - Yoshio Saburi
- Department of Hematology, Oita Prefectural Hospital, Japan
| | - Itaru Matsumura
- Department of Hematology and Rheumatology, Kindai University Faculty of Medicine, Japan
| | - Jun Yamanouchi
- Departments of Hematology, Clinical Immunology and Infectious Diseases, Ehime University Graduate School of Medicine, Japan
| | - Souichi Shiratori
- Department of Hematology, Hokkaido University, Faculty of Medicine, Japan
| | - Moritaka Gotoh
- Department of Hematology, Tokyo Medical University, Japan
| | - Shingen Nakamura
- Department of Community Medicine and Medical Science, Tokushima University Graduate School of Biomedical Sciences, Japan
| | - Kazuo Tamura
- General Medical Research Center, Fukuoka University, Japan
| | | |
Collapse
|
47
|
Hashemian SM, Farhadi T, Velayati AA. Caspofungin: a review of its characteristics, activity, and use in intensive care units. Expert Rev Anti Infect Ther 2020; 18:1213-1220. [PMID: 32662712 DOI: 10.1080/14787210.2020.1794817] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Candidemia is the fourth frequent reason of healthcare-related bloodstream infections in critically ill patients. For initial management of (suspected) invasive candidiasis in critically ill patients, usage of an echinocandin, e.g. caspofungin, has been recommended. AREAS COVERED In this study, characteristics of caspofungin and its use in intensive care unit (ICU) patients are reviewed based on an electronic search using PubMed and Google scholar. EXPERT OPINION Caspofungin is a semisynthetic derivative from pneumocandin B and the first member of the echinocandins that was approved by the U.S. Food and Drug Administration (FDA) to fight fungal infection. Caspofungin inhibits the enzyme β(1,3)-D-glucan synthase of the fungal cell wall resulted in inhibition of the synthesis of β(1,3)-D-glucan. For critically ill patients, inter- and intraindividual variations affect the caspofungin concentration. The incidence rates and densities of candidemia in surgical ICUs may be higher than medical ICUs resulting in a higher burden of candidemia in surgical ICUs. However, the mortality rate in surgical ICU patients with candidemia is higher than that medical ICU patients due to differences in their underlying conditions.
Collapse
Affiliation(s)
- Seyed MohammadReza Hashemian
- Clinical Tuberculosis and Epidemiology Research Center, National Research Institute of Tuberculosis and Lung Disease (NRITLD), Shahid Beheshti University of Medical Sciences , Tehran, Iran.,Chronic Respiratory Diseases Research Center (CRDRC), National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences , Tehran, Iran
| | - Tayebeh Farhadi
- Chronic Respiratory Diseases Research Center (CRDRC), National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences , Tehran, Iran
| | - Ali Akbar Velayati
- Chronic Respiratory Diseases Research Center (CRDRC), National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences , Tehran, Iran
| |
Collapse
|
48
|
Dihydrofolate Reductase Is a Valid Target for Antifungal Development in the Human Pathogen Candida albicans. mSphere 2020; 5:5/3/e00374-20. [PMID: 32581079 PMCID: PMC7316490 DOI: 10.1128/msphere.00374-20] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The folate biosynthetic pathway is a promising and understudied source for novel antifungals. Even dihydrofolate reductase (DHFR), a well-characterized and historically important drug target, has not been conclusively validated as an antifungal target. Here, we demonstrate that repression of DHFR inhibits growth of Candida albicans, a major human fungal pathogen. Methotrexate, an antifolate, also inhibits growth but through pH-dependent activity. In addition, we show that C. albicans has a limited ability to take up or utilize exogenous folates as only the addition of high concentrations of folinic acid restored growth in the presence of methotrexate. Finally, we show that repression of DHFR in a mouse model of infection was sufficient to eliminate host mortality. Our work conclusively establishes DHFR as a valid antifungal target in C. albicans. While the folate biosynthetic pathway has provided a rich source of antibacterial, antiprotozoal, and anticancer therapies, it has not yet been exploited to develop uniquely antifungal agents. Although there have been attempts to develop fungal-specific inhibitors of dihydrofolate reductase (DHFR), the protein itself has not been unequivocally validated as essential for fungal growth or virulence. The purpose of this study was to establish dihydrofolate reductase as a valid antifungal target. Using a strain with doxycycline-repressible transcription of DFR1 (PTETO-DFR1 strain), we were able to demonstrate that Dfr1p is essential for growth in vitro. Furthermore, nutritional supplements of most forms of folate are not sufficient to restore growth when Dfr1p expression is suppressed or when its activity is directly inhibited by methotrexate, indicating that Candida albicans has a limited capacity to acquire or utilize exogenous sources of folate. Finally, the PTETO-DFR1 strain was rendered avirulent in a mouse model of disseminated candidiasis upon doxycycline treatment. Collectively, these results confirm the validity of targeting dihydrofolate reductase and, by inference, other enzymes in the folate biosynthetic pathway as a strategy to devise new and efficacious therapies to combat life-threatening invasive fungal infections. IMPORTANCE The folate biosynthetic pathway is a promising and understudied source for novel antifungals. Even dihydrofolate reductase (DHFR), a well-characterized and historically important drug target, has not been conclusively validated as an antifungal target. Here, we demonstrate that repression of DHFR inhibits growth of Candida albicans, a major human fungal pathogen. Methotrexate, an antifolate, also inhibits growth but through pH-dependent activity. In addition, we show that C. albicans has a limited ability to take up or utilize exogenous folates as only the addition of high concentrations of folinic acid restored growth in the presence of methotrexate. Finally, we show that repression of DHFR in a mouse model of infection was sufficient to eliminate host mortality. Our work conclusively establishes DHFR as a valid antifungal target in C. albicans.
Collapse
|
49
|
Yoshida I, Saito AM, Tanaka S, Choi I, Hidaka M, Miyata Y, Inoue Y, Yamasaki S, Kagoo T, Iida H, Niimi H, Komeno T, Yoshida C, Tajima F, Yamamoto H, Takase K, Ueno H, Shimomura T, Sakai T, Nakashima Y, Yoshida C, Kubonishi S, Sunami K, Yoshida S, Sakurai A, Kaneko Y, Miyazaki Y, Nagai H. Intravenous itraconazole compared with liposomal amphotericin B as empirical antifungal therapy in patients with neutropaenia and persistent fever. Mycoses 2020; 63:794-801. [PMID: 32391919 PMCID: PMC7497187 DOI: 10.1111/myc.13100] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 04/30/2020] [Accepted: 05/04/2020] [Indexed: 12/16/2022]
Abstract
BACKGROUND Fungal infections are a major complication of neutropaenia following chemotherapy. Their early diagnosis is difficult, and empirical antifungal treatment is widely used, and uses of less toxic drugs that reduce breakthrough infection are required. OBJECTIVE We conducted a multicentre, open-label, randomised, non-inferiority trial to compare the safety and efficacy of intravenous itraconazole (ivITCZ) and liposomal amphotericin B (LAmB) as empirical antifungal therapy in patients with haematological malignancies with neutropaenia and persistent fever. METHODS Patients with haematological malignancies who developed fever refractory to broad-spectrum antibacterial agents under neutropaenia conditions were enrolled. Patients were randomised for treatment with LAmB (3.0 mg/kg/d) or ivITCZ (induction: 400 mg/d, maintenance: 200 mg/d). RESULTS Observed overall favourable response rates of 17/52 (32.7%) and 18/50 (36.0%) in the LAmB and ivITCZ groups, with a model-based estimate of a 4% difference (90% CI, -12% to 20%), did not fulfil the statistical non-inferiority criterion. In the LAmB group, there were two cases of breakthrough infection and five cases of probable invasive fungal disease, whereas in the itraconazole group, neither breakthrough infection nor probable invasive fungal disease occurred. Patients in the ivITCZ group had significantly fewer grade 3-4 hypokalaemia-related events than LAmB group patients (P < .01). The overall incidence of adverse events tended to be lower in the ivITCZ group (P = .07). CONCLUSION ivITCZ showed similar efficacy and safety as LAmB as empirical antifungal therapy in haematological malignancy patients with febrile neutropaenia, although the small sample size and various limitations prevented demonstration of its non-inferiority.
Collapse
Affiliation(s)
- Isao Yoshida
- Department of Hematologic Oncology, National Hospital Organization Shikoku Cancer Center, Matsuyama, Japan
| | - Akiko M Saito
- Clinical Research Center, National Hospital Organization Nagoya Medical Center, Nagoya, Japan
| | - Shiro Tanaka
- Department of Clinical Biostatistics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Ilseung Choi
- Department of Hematology, National Hospital Organization Kyushu Cancer Center, Fukuoka, Japan
| | - Michihiro Hidaka
- Department of Hematology, National Hospital Organization Kumamoto Medical Center, Kumamoto, Japan
| | - Yasuhiko Miyata
- Department of Hematology, National Hospital Organization Nagoya Medical Center, Nagoya, Japan
| | - Yoshiko Inoue
- Department of Hematology, National Hospital Organization Kumamoto Medical Center, Kumamoto, Japan
| | - Satoshi Yamasaki
- Department of Hematology and Clinical Research Institute, National Hospital Organization Kyushu Medical Center, Fukuoka, Japan
| | - Toshiya Kagoo
- Department of Hematology and Internal Medicine, National Hospital Organization Tokyo Medical Center, Tokyo, Japan
| | - Hiroatsu Iida
- Department of Hematology, National Hospital Organization Nagoya Medical Center, Nagoya, Japan
| | - Hiromasa Niimi
- Department of Internal Medicine, National Hospital Organization Hiroshima-Nishi Medical Center, Otake, Japan
| | - Takuya Komeno
- Department of Hematology, National Hospital Organization Mito Medical Center, Higashiibarakigun, Japan
| | - Chikamasa Yoshida
- Department of Hematology, National Hospital Organization Minami-Okayama Medical Center, Okayama, Japan
| | - Fumihito Tajima
- Stem Cell Transplantation Center, National Hospital Organization Yonago Medical Center, Yonago, Japan
| | - Hideyuki Yamamoto
- Department of Hematology, National Hospital Organization Nagoya Medical Center, Nagoya, Japan
| | - Ken Takase
- Department of Hematology and Clinical Research Institute, National Hospital Organization Kyushu Medical Center, Fukuoka, Japan
| | - Hironori Ueno
- Department of Hematology and Internal Medicine, National Hospital Organization Tokyo Medical Center, Tokyo, Japan
| | - Takeshi Shimomura
- Department of Internal Medicine, National Hospital Organization Hiroshima-Nishi Medical Center, Otake, Japan
| | - Tatsunori Sakai
- Department of Hematology, National Hospital Organization Kumamoto Medical Center, Kumamoto, Japan
| | - Yasuhiro Nakashima
- Department of Hematology, National Hospital Organization Kyushu Cancer Center, Fukuoka, Japan
| | - Chikashi Yoshida
- Department of Hematology, National Hospital Organization Mito Medical Center, Higashiibarakigun, Japan
| | - Shiro Kubonishi
- Department of Hematology, National Hospital Organization Okayama Medical Center, Okayama, Japan
| | - Kazutaka Sunami
- Department of Hematology, National Hospital Organization Okayama Medical Center, Okayama, Japan
| | - Shinichiro Yoshida
- Department of Hematology, National Hospital Organization Nagasaki Medical Center, Omura, Japan
| | - Aki Sakurai
- Department of Hematologic Oncology, National Hospital Organization Shikoku Cancer Center, Matsuyama, Japan
| | - Yukihiro Kaneko
- Department of Bacteriology, Osaka City University Graduate School of Medicine, Osaka, Japan.,Department of Chemotherapy and Mycoses, National Institute of Infectious Diseases, Tokyo, Japan
| | - Yoshitsugu Miyazaki
- Department of Chemotherapy and Mycoses, National Institute of Infectious Diseases, Tokyo, Japan
| | - Hirokazu Nagai
- Department of Hematology, National Hospital Organization Nagoya Medical Center, Nagoya, Japan
| |
Collapse
|
50
|
Tori K, Tansarli GS, Parente DM, Kalligeros M, Ziakas PD, Mylonakis E. The cost-effectiveness of empirical antibiotic treatments for high-risk febrile neutropenic patients: A decision analytic model. Medicine (Baltimore) 2020; 99:e20022. [PMID: 32443305 PMCID: PMC7254453 DOI: 10.1097/md.0000000000020022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
PURPOSE Febrile neutropenia has a significant clinical and economic impact on cancer patients. This study evaluates the cost-effectiveness of different current empiric antibiotic treatments. METHODS A decision analytic model was constructed to compare the use of cefepime, meropenem, imipenem/cilastatin, and piperacillin/tazobactam for treatment of high-risk patients. The analysis was performed from the perspective of U.S.-based hospitals. The time horizon was defined to be a single febrile neutropenia episode. Cost-effectiveness was determined by calculating costs and deaths averted. Cost-effectiveness acceptability curves for various willingness-to-pay thresholds (WTP), were used to address the uncertainty in cost-effectiveness. RESULTS The base-case analysis results showed that treatments were equally effective but differed mainly in their cost. In increasing order: treatment with imipenem/cilastatin cost $52,647, cefepime $57,270, piperacillin/tazobactam $57,277, and meropenem $63,778. In the probabilistic analysis, mean costs were $52,554 (CI: $52,242-$52,866) for imipenem/cilastatin, $57,272 (CI: $56,951-$57,593) for cefepime, $57,294 (CI: $56,978-$57,611) for piperacillin/tazobactam, and $63,690 (CI: $63,370-$64,009) for meropenem. Furthermore, with a WTP set at $0 to $50,000, imipenem/cilastatin was cost-effective in 66.2% to 66.3% of simulations compared to all other high-risk options. DISCUSSION Imipenem/cilastatin is a cost-effective strategy and results in considerable health care cost-savings at various WTP thresholds. Cost-effectiveness analyses can be used to differentiate the treatments of febrile neutropenia in high-risk patients.
Collapse
Affiliation(s)
- Katerina Tori
- Division of Infectious Diseases, Brown University, Warren Alpert Medical School
| | | | - Diane M. Parente
- Department of Pharmacy, The Miriam Hospital, Providence, Rhode Island, USA
| | - Markos Kalligeros
- Division of Infectious Diseases, Brown University, Warren Alpert Medical School
| | | | | |
Collapse
|