1
|
Wang M, Yang Y, Li D, Wang Y, Ji T, Li Q, Zhang J, Zhang P, Su J. Miconazole-splitomicin combined β-glucan hydrogel for effective prevention of Candida albicans Periprosthetic Joint Infection. Eur J Pharm Sci 2024:106955. [PMID: 39505047 DOI: 10.1016/j.ejps.2024.106955] [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: 05/07/2024] [Revised: 11/01/2024] [Accepted: 11/03/2024] [Indexed: 11/08/2024]
Abstract
As one of the most common and serious infections caused by Candida albicans (C. albicans), periprosthetic joint infection (PJI) increasingly concerns surgeons and scientists. Generally, biofilms shield C. albicans from antifungal agents and immune clearance and induce drug-resistant strains. Developing novel strategies for PJI to get rid of current drug-resistant problems is highly needed. In our study, splitomicin (SP) can inhibit the mycelium formation of C. albicans and enhance the drug sensitivity of C. albicans to miconazole nitrate (MCZ). The combination of SP and MCZ significantly inhibited the viability, proliferation and adhesion of C. albicans, reduced the yeast to hyphae transition and biofilm formation. When SP and MCZ were coloaded in the β-glucan hydrogel, a viscoelastic solid with porous 3D network, sustained release and erosion properties was obtained. In the in vivo PJI mice model, SP-MCZ-β-glucan hydrogel effectively reduced the colonization and aggregation of C. albicans around the implant, reduced the pathological changes caused by C. albicans in the femur tissue. Therefore, SP-MCZ-β-glucan hydrogel holds a great promise for the management of C. albicans infection around joint prosthesis.
Collapse
Affiliation(s)
- Menghan Wang
- The first affiliated hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P. R. China; School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, P. R. China
| | - Ying Yang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, P. R. China
| | - Dongdong Li
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, P. R. China
| | - Yanmei Wang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, P. R. China
| | - Tailin Ji
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, P. R. China
| | - Qingqing Li
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, P. R. China
| | - Jiye Zhang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, P. R. China
| | - Peipei Zhang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, P. R. China.
| | - Jin Su
- The first affiliated hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P. R. China.
| |
Collapse
|
2
|
Yeoh DK, Blyth CC, Clark JE, Abbotsford J, Corrente C, Cook S, Kotecha RS, Wang SS, Spelman T, Slavin MA, Thursky KA, Haeusler GM. Invasive fungal disease and antifungal prophylaxis in children with acute leukaemia: a multicentre retrospective Australian cohort study. THE LANCET REGIONAL HEALTH. WESTERN PACIFIC 2024; 52:101201. [PMID: 39318715 PMCID: PMC11417227 DOI: 10.1016/j.lanwpc.2024.101201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 07/26/2024] [Accepted: 08/28/2024] [Indexed: 09/26/2024]
Abstract
Background Invasive fungal disease (IFD) is a significant complication for children receiving treatment for leukaemia, contributing to morbidity and mortality. Recent regional paediatric epidemiological IFD data are lacking. Additionally uncertainty remains regarding the optimal prophylactic approach in this context. Methods In a multi-centre Australian cohort study of children diagnosed with de novo acute leukaemia between 1st January 2017 and 30th June 2020, we characterised antifungal prophylaxis prescribing and IFD prevalence. Impact of antifungal prophylaxis was assessed using Kaplan Meier curves and Cox-proportional hazards regression adjusting for known IFD risk factors. Findings A total of 434 children were included (47.2% female; median age 5.0 years, median follow-up 240 days). This cohort included 351 children with ALL (214 high-risk [HR-ALL]; 137 standard-risk [SR-ALL]), and 73 with AML. The prevalence of proven/probable IFD was 6.8% for AML, 14.0% for HR-ALL and 4.4% for SR-ALL. A mould was implicated as the causative pathogen in almost two thirds of cases. Antifungal prophylaxis was prescribed in 98.7% of chemotherapy cycles for AML, 56.7% for HR-ALL and 14.9% for SR-ALL. A mould-active agent was used in 77.4% of AML cycles and 21.2% of HR-ALL cycles. Mould-active prophylaxis was associated with a lower risk of IFD overall and increased IFD-free survival in AML. Interpretation These data demonstrate the persistent high regional burden of IFD in children with HR-ALL, and the potential for mould-active prophylaxis to ameliorate this. Strategies to increase uptake of appropriate prophylaxis are required in this cohort. Funding This study was supported by a Perth Children's Hospital Foundation grant (PCHF9973).
Collapse
Affiliation(s)
- Daniel K. Yeoh
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
- Department of Infectious Diseases, Perth Children's Hospital, Perth, Australia
- National Centre for Infections in Cancer, Peter MacCallum Cancer Centre, Melbourne, Australia
- Murdoch Children's Research Institute, Melbourne, Australia
- Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, University of Western Australia, Perth, Australia
| | - Christopher C. Blyth
- Department of Infectious Diseases, Perth Children's Hospital, Perth, Australia
- Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, University of Western Australia, Perth, Australia
- Department of Microbiology, PathWest Laboratory Medicine WA, Perth, Australia
- School of Medicine, University of Western Australia, Perth, Australia
| | - Julia E. Clark
- Infection Management Service, Queensland Children's Hospital, Brisbane, Australia
- School of Clinical Medicine, Children's Health Queensland Clinical Unit, The University of Queensland, Brisbane, Australia
| | - Joanne Abbotsford
- Department of Infectious Diseases, Perth Children's Hospital, Perth, Australia
| | | | - Sara Cook
- Infection Management Service, Queensland Children's Hospital, Brisbane, Australia
| | - Rishi S. Kotecha
- Department of Clinical Haematology, Oncology, Blood and Marrow Transplantation, Perth Children's Hospital, Perth, Australia
- Curtin Medical School, Curtin University, Perth, Australia
- Leukaemia Translational Research Laboratory, Telethon Kids Cancer Centre, Telethon Kids Institute, University of Western Australia, Perth, Australia
| | - Stacie S. Wang
- Murdoch Children's Research Institute, Melbourne, Australia
- Children's Cancer Centre, Royal Children's Hospital, Melbourne, Australia
| | - Tim Spelman
- National Centre for Infections in Cancer, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Monica A. Slavin
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
- National Centre for Infections in Cancer, Peter MacCallum Cancer Centre, Melbourne, Australia
- Department of Infectious Diseases, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Karin A. Thursky
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
- National Centre for Infections in Cancer, Peter MacCallum Cancer Centre, Melbourne, Australia
- Department of Infectious Diseases, Peter MacCallum Cancer Centre, Melbourne, Australia
- National Centre for Antimicrobial Stewardship, Department of Infectious Diseases, Faculty of Medicine Dentistry and Health Sciences, University of Melbourne, Melbourne, Australia
| | - Gabrielle M. Haeusler
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
- National Centre for Infections in Cancer, Peter MacCallum Cancer Centre, Melbourne, Australia
- Murdoch Children's Research Institute, Melbourne, Australia
- Department of Infectious Diseases, Peter MacCallum Cancer Centre, Melbourne, Australia
- Department of Infectious Diseases, Royal Children's Hospital, Melbourne, Australia
- The Paediatric Integrated Cancer Service, Melbourne, Australia
| |
Collapse
|
3
|
Yeoh DK, Haeusler GM, Slavin MA, Kotecha RS. Challenges and considerations for antifungal prophylaxis in children with acute myeloid leukemia. Expert Rev Hematol 2024; 17:679-686. [PMID: 39110722 DOI: 10.1080/17474086.2024.2390639] [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: 04/16/2024] [Accepted: 08/06/2024] [Indexed: 09/21/2024]
Abstract
INTRODUCTION Children receiving treatment for acute myeloid leukemia (AML) are at high risk of invasive fungal disease (IFD). Evidence from pediatric studies support the efficacy of antifungal prophylaxis in reducing the burden of IFD in children receiving therapy for AML, yet existing antifungal agents have specific limitations and comparative data to inform the optimal prophylactic approach are lacking. AREAS COVERED This review summarizes the epidemiology of invasive fungal disease (IFD) and current antifungal prophylaxis recommendations for children with acute myeloid leukemia (AML). Challenges with currently available antifungal agents and considerations related to the changing landscape of AML therapy are reviewed. A keyword search was conducted to identify pediatric studies regarding IFD and antifungal prophylaxis in children with AML up to December 2023. EXPERT OPINION Children undergoing treatment for AML are recommended to receive antifungal prophylaxis to reduce risk of IFD, with tolerability, pharmacokinetics, feasibility of administration, and drug interactions all factors that require consideration in this context. With increased use of novel targeted agents for AML therapy, together with the development of new antifungal agents, data from well-designed clinical studies to optimize prophylactic approaches will be essential to limit the burden of IFD in this vulnerable cohort.
Collapse
Affiliation(s)
- Daniel K Yeoh
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
- Department of Infectious Diseases, Perth Children's Hospital, Perth, Australia
- National Centre for Infections in Cancer, Peter MacCallum Cancer Centre, Melbourne, Australia
- Infection, Immunity and Global Health, Murdoch Children's Research Institute, Melbourne, Australia
- Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, University of Western Australia, Perth, Australia
| | - Gabrielle M Haeusler
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
- National Centre for Infections in Cancer, Peter MacCallum Cancer Centre, Melbourne, Australia
- Infection, Immunity and Global Health, Murdoch Children's Research Institute, Melbourne, Australia
- Department of Infectious Diseases, Peter MacCallum Cancer Centre, Melbourne, Australia
- Department of Infectious Diseases, Royal Children's Hospital, Melbourne, Australia
| | - Monica A Slavin
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
- National Centre for Infections in Cancer, Peter MacCallum Cancer Centre, Melbourne, Australia
- Department of Infectious Diseases, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Rishi S Kotecha
- Department of Clinical Haematology, Oncology, Blood and Marrow Transplantation, Perth Children's Hospital, Perth, Australia
- Curtin Medical School, Curtin University, Perth, Australia
- Leukaemia Translational Research Laboratory, Telethon Kids Cancer Centre, Telethon Kids Institute, University of Western Australia, Perth, Australia
| |
Collapse
|
4
|
Groll AH, Körholz K, Holterhus M, Lehrnbecher T. New and emerging options for management of invasive fungal diseases in paediatric patients. Mycoses 2024; 67:e13654. [PMID: 37789721 DOI: 10.1111/myc.13654] [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: 07/10/2023] [Revised: 08/30/2023] [Accepted: 09/02/2023] [Indexed: 10/05/2023]
Abstract
Invasive fungal diseases (IFDs) play an important role in the supportive care of paediatric patients with acute leukaemia and those undergoing allogeneic haematopoietic cell transplantation, and they are associated with significantly decreased overall survival rates in affected individuals. Relative to adults, children and adolescents are distinct in terms of host biology, predisposing conditions, presentation and epidemiology of fungal diseases, and in the pharmacology of antifungal agents. The paediatric development of antifungal agents has moved forward in a coordinated manner, and major advances have been made regarding concepts and recommendations for the prevention and treatment of IFDs. However, antifungal therapy is increasingly complex, and a solid knowledge of the available options is needed more than ever for successful management. This narrative review provides a summary of the paediatric development of agents that have been recently approved (anidulafungin, posaconazole) or are in advanced stages of development (isavuconazole). It also reviews the emerging evidence for the efficacy of echinocandins for prophylaxis of invasive aspergillosis, presents new data on alternative dosing regimens of echinocandins and voriconazole, and provides a brief overview of new antifungal agents in clinical development that are expected to be developed for paediatric patients.
Collapse
Affiliation(s)
- Andreas H Groll
- Infectious Disease Research Program, Center for Bone Marrow Transplantation and Department of Pediatric Hematology/Oncology, Children's University Hospital Münster, Münster, Germany
| | - Katharina Körholz
- Infectious Disease Research Program, Center for Bone Marrow Transplantation and Department of Pediatric Hematology/Oncology, Children's University Hospital Münster, Münster, Germany
| | - Malcolm Holterhus
- Infectious Disease Research Program, Center for Bone Marrow Transplantation and Department of Pediatric Hematology/Oncology, Children's University Hospital Münster, Münster, Germany
| | - Thomas Lehrnbecher
- Department of Pediatrics, Division of Hematology, Oncology and Hemostaseology, Goethe University Frankfurt, Frankfurt, Germany
| |
Collapse
|
5
|
Groll AH, Ebrahimi-Fakhari D. Conference Report 33rd European Congress on Clinical Microbiology and Infectious Diseases (ECCMID): New developments in pediatric oncology infectious disease supportive care. Transpl Infect Dis 2023; 25:e14146. [PMID: 37695128 DOI: 10.1111/tid.14146] [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: 08/25/2023] [Accepted: 08/27/2023] [Indexed: 09/12/2023]
Abstract
Infections continue to be major causes of morbidity and mortality in immunocompromised children and adolescents with cancer or undergoing allogeneic hematopoietic cell transplantation. This report summarizes new clinical research data presented at the 33rd European Congress on Clinical Microbiology and Infectious Diseases on infections in this vulnerable population, with a focus on the epidemiology, diagnosis, and prevention of invasive fungal diseases and de-escalation strategies in neutropenic patients with fever of unknown origin.
Collapse
Affiliation(s)
- Andreas H Groll
- Infectious Disease Research Program, Center for Bone Marrow Transplantation and Department of Pediatric Hematology and Oncology, University Children´s Hospital Münster, Münster, Germany
| | - Daniel Ebrahimi-Fakhari
- Infectious Disease Research Program, Center for Bone Marrow Transplantation and Department of Pediatric Hematology and Oncology, University Children´s Hospital Münster, Münster, Germany
| |
Collapse
|
6
|
Lehrnbecher T, Bochennek K, Groll AH. Mold-Active Antifungal Prophylaxis in Pediatric Patients with Cancer or Undergoing Hematopoietic Cell Transplantation. J Fungi (Basel) 2023; 9:387. [PMID: 36983555 PMCID: PMC10059906 DOI: 10.3390/jof9030387] [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: 03/02/2023] [Revised: 03/18/2023] [Accepted: 03/21/2023] [Indexed: 03/30/2023] Open
Abstract
Invasive fungal diseases (IFDs), in particular invasive mold infections, still pose considerable problems in the care of children and adolescents treated for cancer or undergoing hematopoietic cell transplantation. As these infections are difficult to diagnose, and the outcomes for IFDs are still unsatisfactory, antifungal prophylaxis has become an important strategy in the clinical setting. Antifungal prophylaxis is indicated in patients at high risk for IFD, which is commonly defined as a natural incidence of at least 10%. As there is a growing interest in pediatric-specific clinical trials and pediatric-specific guidelines, this review focuses on the available data of mold-active antifungal prophylaxis in children and adolescents. The data demonstrate that a major effort is needed to characterize the pediatric patient population in which the net effect of prophylactic antifungals will be beneficial as well as to find the optimal prophylactic antifungal compound and dosage.
Collapse
Affiliation(s)
- Thomas Lehrnbecher
- Division of Pediatric Hematology and Oncology, Hospital for Children and Adolescents, University Hospital, Johann Wolfgang Goethe University, 60589 Frankfurt am Main, Germany
| | - Konrad Bochennek
- Division of Pediatric Hematology and Oncology, Hospital for Children and Adolescents, University Hospital, Johann Wolfgang Goethe University, 60589 Frankfurt am Main, Germany
| | - Andreas H. Groll
- Infectious Disease Research Program, Center for Bone Marrow Transplantation, Department of Pediatric Hematology/Oncology, University Children’s Hospital, 48149 Muenster, Germany
| |
Collapse
|
7
|
Haro-Reyes T, Díaz-Peralta L, Galván-Hernández A, Rodríguez-López A, Rodríguez-Fragoso L, Ortega-Blake I. Polyene Antibiotics Physical Chemistry and Their Effect on Lipid Membranes; Impacting Biological Processes and Medical Applications. MEMBRANES 2022; 12:681. [PMID: 35877884 PMCID: PMC9316096 DOI: 10.3390/membranes12070681] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 06/21/2022] [Accepted: 06/23/2022] [Indexed: 01/27/2023]
Abstract
This review examined a collection of studies regarding the molecular properties of some polyene antibiotic molecules as well as their properties in solution and in particular environmental conditions. We also looked into the proposed mechanism of action of polyenes, where membrane properties play a crucial role. Given the interest in polyene antibiotics as therapeutic agents, we looked into alternative ways of reducing their collateral toxicity, including semi-synthesis of derivatives and new formulations. We follow with studies on the role of membrane structure and, finally, recent developments regarding the most important clinical applications of these compounds.
Collapse
Affiliation(s)
- Tammy Haro-Reyes
- Instituto de Ciencias Físicas, Universidad Nacional Autónoma de México, Av. Universidad s/n, Col. Chamilpa, Cuernavaca 62210, Morelos, Mexico; (T.H.-R.); (L.D.-P.); (A.G.-H.)
| | - Lucero Díaz-Peralta
- Instituto de Ciencias Físicas, Universidad Nacional Autónoma de México, Av. Universidad s/n, Col. Chamilpa, Cuernavaca 62210, Morelos, Mexico; (T.H.-R.); (L.D.-P.); (A.G.-H.)
| | - Arturo Galván-Hernández
- Instituto de Ciencias Físicas, Universidad Nacional Autónoma de México, Av. Universidad s/n, Col. Chamilpa, Cuernavaca 62210, Morelos, Mexico; (T.H.-R.); (L.D.-P.); (A.G.-H.)
| | - Anahi Rodríguez-López
- Facultad de Farmacia, Universidad Autónoma del Estado de Morelos, Cuernavaca 62210, Morelos, Mexico; (A.R.-L.); (L.R.-F.)
| | - Lourdes Rodríguez-Fragoso
- Facultad de Farmacia, Universidad Autónoma del Estado de Morelos, Cuernavaca 62210, Morelos, Mexico; (A.R.-L.); (L.R.-F.)
| | - Iván Ortega-Blake
- Instituto de Ciencias Físicas, Universidad Nacional Autónoma de México, Av. Universidad s/n, Col. Chamilpa, Cuernavaca 62210, Morelos, Mexico; (T.H.-R.); (L.D.-P.); (A.G.-H.)
| |
Collapse
|
8
|
Gastine S, Hempel G, Neely MN, Walsh TJ, Groll AH. Pharmacokinetic modelling of caspofungin to develop an extended dosing regimen in paediatric patients. J Antimicrob Chemother 2022; 77:2209-2216. [PMID: 35696555 DOI: 10.1093/jac/dkac182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 05/16/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Echinocandins are commonly used in treatment and prophylaxis of invasive fungal diseases. Intravenous daily dosing for prophylaxis in the outpatient setting can however become a hurdle for adequate compliance in the paediatric population. OBJECTIVES Simulations were performed to assess extended twice-weekly dosing for antifungal prophylaxis using caspofungin. METHODS A population pharmacokinetic model was developed based on previously published data from children aged 3 months to 17 years. Using the final model, Monte Carlo simulations were performed to assess the dose needed for adequate exposure in a twice-weekly setting. Mean weekly AUC0-24 h/MIC together with reported AUC0-24 h from previously reported paediatric trials were used to guide adequate exposure. RESULTS AND CONCLUSIONS A two-compartment model with linear elimination and allometric scaling using fixed exponents was found most adequate to describe the given paediatric populations. Simulations showed that a 200 mg/m2 twice-weekly regimen with maximal 200 mg total dose should result in exposures matching registered daily dosing as well as commonly used pharmacokinetic/pharmacodynamic targets.
Collapse
Affiliation(s)
- Silke Gastine
- Institute of Pharmaceutical and Medical Chemistry, Deptartment of Clinical Pharmacy, Westfälische Wilhelms-Universität Münster, Münster, Germany.,Department of Infection, Immunity & Inflammation Research & Teaching, GOS Institute of Child Health, University College London, London, UK
| | - Georg Hempel
- Institute of Pharmaceutical and Medical Chemistry, Deptartment of Clinical Pharmacy, Westfälische Wilhelms-Universität Münster, Münster, Germany
| | - Michael N Neely
- Division of Infectious Diseases, Department of Pediatrics, Children's Hospital Los Angeles, Los Angeles, CA, USA
| | - Thomas J Walsh
- Transplantation-Oncology Infectious Diseases Program, Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine of Cornell University, New York, NY, USA
| | - Andreas H Groll
- Infectious Disease Research Program, Center for Bone Marrow Transplantation and Dept. of Pediatric Hematology/Oncology, University Children's Hospital Münster, Münster, Germany
| |
Collapse
|
9
|
|
10
|
Management of drug–drug interactions of targeted therapies for haematological malignancies and triazole antifungal drugs. THE LANCET HAEMATOLOGY 2022; 9:e58-e72. [DOI: 10.1016/s2352-3026(21)00232-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 07/01/2021] [Accepted: 07/19/2021] [Indexed: 12/11/2022]
|
11
|
Teh BW, Yeoh DK, Haeusler GM, Yannakou CK, Fleming S, Lindsay J, Slavin MA. Consensus guidelines for antifungal prophylaxis in haematological malignancy and haemopoietic stem cell transplantation, 2021. Intern Med J 2021; 51 Suppl 7:67-88. [PMID: 34937140 DOI: 10.1111/imj.15588] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Antifungal prophylaxis can reduce morbidity and mortality from invasive fungal disease (IFD). However, its use needs to be optimised and appropriately targeted to patients at highest risk to derive the most benefit. In addition to established risks for IFD, considerable recent progress in the treatment of malignancies has resulted in the development of new 'at-risk' groups. The changing epidemiology of IFD and emergence of drug resistance continue to impact choice of prophylaxis, highlighting the importance of active surveillance and knowledge of local epidemiology. These guidelines aim to highlight emerging risk groups and review the evidence and limitations around new formulations of established agents and new antifungal drugs. It provides recommendations around use and choice of antifungal prophylaxis, discusses the potential impact of the changing epidemiology of IFD and emergence of drug resistance, and future directions for risk stratification to assist optimal management of highly vulnerable patients.
Collapse
Affiliation(s)
- Benjamin W Teh
- Department of Infectious Diseases, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia.,National Centre for Infections in Cancer, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Daniel K Yeoh
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia.,National Centre for Infections in Cancer, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Department of Infectious Diseases, Perth Children's Hospital, Perth, Western Australia, Australia
| | - Gabrielle M Haeusler
- Department of Infectious Diseases, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia.,National Centre for Infections in Cancer, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Department of Infectious Diseases, Royal Children's Hospital, Melbourne, Victoria, Australia.,Murdoch Children's Research Institute, Parkville, Victoria, Australia
| | - Costas K Yannakou
- Department of Molecular Oncology and Cancer Immunology, Epworth Freemasons Hospital, Epworth HealthCare, Melbourne, Victoria, Australia
| | - Shaun Fleming
- Malignant Haematology and Stem Cell Transplantation Service, Alfred Health, Melbourne, Victoria, Australia
| | - Julian Lindsay
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia.,National Centre for Infections in Cancer, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Department of Haematology, Royal North Shore Hospital, Sydney, New South Wales, Australia
| | - Monica A Slavin
- Department of Infectious Diseases, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia.,National Centre for Infections in Cancer, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Immunocompromised Host Infection Service, Royal Melbourne Hospital, Melbourne, Victoria, Australia
| | | |
Collapse
|
12
|
Yeoh DK, Moore AS, Kotecha RS, Bartlett AW, Ryan AL, Cann MP, McMullan BJ, Thursky K, Slavin M, Blyth CC, Haeusler GM, Clark JE. Invasive fungal disease in children with acute myeloid leukaemia: An Australian multicentre 10-year review. Pediatr Blood Cancer 2021; 68:e29275. [PMID: 34357688 DOI: 10.1002/pbc.29275] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 07/01/2021] [Accepted: 07/16/2021] [Indexed: 12/19/2022]
Abstract
BACKGROUND Invasive fungal disease (IFD) is a common and important complication in children with acute myeloid leukaemia (AML). We describe the epidemiology of IFD in a large multicentre cohort of children with AML. METHODS As part of the retrospective multicentre cohort TERIFIC (The Epidemiology and Risk factors for Invasive Fungal Infections in immunocompromised Children) study, proven/probable/possible IFD episodes occurring in children with primary or relapsed/refractory AML from 2003 to 2014 were analysed. Crude IFD prevalence, clinical characteristics, microbiology and treatment were assessed. Kaplan-Meier survival analysis was used to estimate 6-month survival. RESULTS There were 66 IFD episodes diagnosed in 63 children with AML. The majority (75.8%) of episodes occurred in the context of primary AML therapy. During primary AML therapy, the overall prevalence was 20.7% (95% CI 15.7%-26.5%) for proven/probable/possible IFD and 10.3% (95% CI 6.7%-15.0%) for proven/probable IFD. Of primary AML patients, 8.2% had IFD diagnosed during the first cycle of chemotherapy. Amongst pathogens implicated in proven/probable IFD episodes, 74.4% were moulds, over a third (37.9%) of which were non-Aspergillus spp. Antifungal prophylaxis preceded 89.4% of IFD episodes, most commonly using fluconazole (50% of IFD episodes). All-cause mortality at 6 months from IFD diagnosis was 16.7% with IFD-related mortality of 7.6% (all in cases of proven IFD). CONCLUSIONS IFD is a common and serious complication during paediatric AML therapy. Mould infections, including non-Aspergillus spp. predominated in this cohort. A systematic approach to the identification of patients at risk, and a targeted prevention strategy for IFD is needed.
Collapse
Affiliation(s)
- Daniel K Yeoh
- Department of Infectious Diseases, Perth Children's Hospital, Perth, Western Australia, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia.,National Centre for Infections in Cancer, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Murdoch Children's Research Institute, Parkville, Victoria, Australia
| | - Andrew S Moore
- Oncology Service, Queensland Children's Hospital, Brisbane, Queensland, Australia.,Child Health Research Centre, The University of Queensland, Brisbane, Queensland, Australia
| | - Rishi S Kotecha
- Department of Clinical Haematology, Oncology, Blood and Marrow Transplantation, Perth Children's Hospital, Perth, Western Australia, Australia.,Curtin Medical School, Curtin University, Perth, Western Australia, Australia.,Telethon Kids Cancer Centre, Telethon Kids Institute, University of Western, Perth, Western Australia, Australia
| | - Adam W Bartlett
- Department of Immunology and Infectious Diseases, Sydney Children's Hospital, Randwick, New South Wales, Australia.,School of Women's and Children's Health, UNSW, Sydney, New South Wales, Australia.,Kirby Institute, UNSW, Sydney, New South Wales, Australia
| | - Anne L Ryan
- Curtin Medical School, Curtin University, Perth, Western Australia, Australia
| | - Megan P Cann
- Infection Management Service, Queensland Children's Hospital, Brisbane, Queensland, Australia
| | - Brendan J McMullan
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia.,National Centre for Infections in Cancer, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Department of Immunology and Infectious Diseases, Sydney Children's Hospital, Randwick, New South Wales, Australia.,School of Women's and Children's Health, UNSW, Sydney, New South Wales, Australia
| | - Karin Thursky
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia.,National Centre for Infections in Cancer, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Department of Infectious Diseases, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,National Health and Medical Research Council National Centre for Antimicrobial Stewardship, The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Monica Slavin
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia.,National Centre for Infections in Cancer, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Department of Infectious Diseases, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Christopher C Blyth
- Department of Infectious Diseases, Perth Children's Hospital, Perth, Western Australia, Australia.,Department of Microbiology, PathWest Laboratory Medicine, Nedlands, Western Australia, Australia.,Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, University of Western, Perth, Western Australia, Australia.,School of Medicine, University of Western Australia, Perth, Western Australia, Australia
| | - Gabrielle M Haeusler
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia.,National Centre for Infections in Cancer, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Murdoch Children's Research Institute, Parkville, Victoria, Australia.,Department of Infectious Diseases, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Department of Infectious Diseases, Royal Children's Hospital, Parkville, Victoria.,The Paediatric Integrated Cancer Service, Melbourne, Victoria, Australia
| | - Julia E Clark
- Infection Management Service, Queensland Children's Hospital, Brisbane, Queensland, Australia.,School of Clinical Medicine, Children's Health Queensland Clinical Unit, The University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|
13
|
Sheng B, Chen Y, Sun L, Xu P, Han B, Li X, Yin J, Li T, Guan H, Chen S, Wang Q, Li C, Li S, Jiang X, Wang P, He Q, Wang Y, Xiao W, Yang H. Antifungal Treatment Aggravates Sepsis through the Elimination of Intestinal Fungi. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:2796700. [PMID: 34707775 PMCID: PMC8545547 DOI: 10.1155/2021/2796700] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Accepted: 09/27/2021] [Indexed: 12/26/2022]
Abstract
Prophylactic antifungal therapy is widely adopted clinically for critical patients and effective in reducing the morbidity of invasive fungal infection and improves outcomes of those diagnosed patients; however, it is not associated with higher overall survival. As intestinal commensal fungi play a fundamental role in the host immune response in health and disease, we propose that antifungal therapy may eliminate intestinal fungi and aggravate another critical syndrome, sepsis. Here, with murine sepsis model, we found that antifungal therapy with fluconazole dismissed intestinal fungal burden and aggravated endotoxin-induced but no gram-positive bacteria-induced sepsis. Nevertheless, antifungal therapy did not exert its detrimental effect on germ-free mice. Moreover, colonizing more commensal fungi in the mouse intestine or administration of fungal cell wall component mannan protected the mice from endotoxin-induced sepsis. On the molecular level, we demonstrated that antifungal therapy aggravated endotoxin sepsis through promoting Gasdermin D cleavage in the distal small intestine. Intestinal colonization with commensal fungi inhibited Gasdermin D cleavage in response to lipopolysaccharide challenge. These findings show that intestinal fungi inhibit Gasdermin D-mediated pyroptosis and protect the mice from endotoxin-induced sepsis. This study demonstrates the protective role of intestinal fungi in the pathogenesis of endotoxin-induced sepsis in the laboratory. It will undoubtedly prompt us to study the relationship between antifungal therapy and sepsis in critical patients who are susceptible to endotoxin-induced sepsis in the future.
Collapse
Affiliation(s)
- Baifa Sheng
- Department of General Surgery, Xinqiao Hospital, Army Medical University, Chongqing 400037, China
- Department of Emergency Medicine, PLA Strategic Support Force Characteristic Medical Center, Beijing 100101, China
| | - Yihui Chen
- Department of General Surgery, Xinqiao Hospital, Army Medical University, Chongqing 400037, China
- Department of General Surgery, The First Mobile Corps Hospital of PAP, Dingzhou City, 073000 Hebei Province, China
| | - Lihua Sun
- Department of General Surgery, Xinqiao Hospital, Army Medical University, Chongqing 400037, China
| | - Peng Xu
- Department of General Surgery, Xinqiao Hospital, Army Medical University, Chongqing 400037, China
| | - Ben Han
- Department of Nutrition, Xinqiao Hospital, Army Medical University, Chongqing 400037, China
| | - Xiaolong Li
- Department of General Surgery, Xinqiao Hospital, Army Medical University, Chongqing 400037, China
| | - Jiuheng Yin
- Department of General Surgery, Xinqiao Hospital, Army Medical University, Chongqing 400037, China
| | - Teming Li
- Department of General Surgery, Xinqiao Hospital, Army Medical University, Chongqing 400037, China
| | - Haidi Guan
- Department of General Surgery, Xinqiao Hospital, Army Medical University, Chongqing 400037, China
| | - Shuaishuai Chen
- Department of General Surgery, Xinqiao Hospital, Army Medical University, Chongqing 400037, China
| | - Qi Wang
- Division of Hematology-Oncology, Department of Medicine Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Chuangen Li
- Department of Laboratory Animal Science, College of Basic Medical Science, Army Medical University, Chongqing 400038, China
| | - Shiqiang Li
- Department of Laboratory Animal Science, College of Basic Medical Science, Army Medical University, Chongqing 400038, China
| | - Xianhong Jiang
- Department of Laboratory Animal Science, College of Basic Medical Science, Army Medical University, Chongqing 400038, China
| | - Peng Wang
- Department of Laboratory Animal Science, College of Basic Medical Science, Army Medical University, Chongqing 400038, China
| | - Qiuyue He
- Department of Laboratory Animal Science, College of Basic Medical Science, Army Medical University, Chongqing 400038, China
| | - Yong Wang
- Department of Laboratory Animal Science, College of Basic Medical Science, Army Medical University, Chongqing 400038, China
| | - Weidong Xiao
- Department of General Surgery, Xinqiao Hospital, Army Medical University, Chongqing 400037, China
| | - Hua Yang
- Department of General Surgery, Xinqiao Hospital, Army Medical University, Chongqing 400037, China
| |
Collapse
|
14
|
van de Peppel RJ, Schauwvlieghe A, Van Daele R, Spriet I, Van't Wout JW, Brüggemann RJ, Rijnders BJA, Hendriks BJC, de Boer MGJ. Outpatient parenteral antifungal therapy (OPAT) for invasive fungal infections with intermittent dosing of liposomal amphotericin B. Med Mycol 2021; 58:874-880. [PMID: 31965178 PMCID: PMC7527269 DOI: 10.1093/mmy/myz134] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 12/10/2019] [Accepted: 12/20/2019] [Indexed: 12/27/2022] Open
Abstract
Triazole resistant A. fumigatus has been documented in many parts of the world. In the Netherlands, incidence is now above 10% and results in the need for long-term parenteral therapy with liposomal amphotericin B (LAmB). The long terminal half-life of LAmB suggests that intermittent dosing could be effective, making the application of outpatient antifungal therapy (OPAT) possible. Here, we report our experience with the use of OPAT for Invasive Fungal Infections (IFI). All adult patients treated with LAmB with a 2 or 3 times weekly administration via the outpatient departments in four academic tertiary care centers in the Netherlands and Belgium since January 2010 were included in our analysis. Patient characteristics were collected, as well as information about diagnostics, therapy dose and duration, toxicity, treatment history and outcome of the IFI. In total, 18 patients were included. The most frequently used regimen (67%) was 5 mg/kg 3 times weekly. A partial response to the daily treatment prior to discharge was confirmed by CT-scan in 17 (94%) of patients. A favorable outcome was achieved in 13 (72%) patients. Decrease in renal function occurred in 10 (56%) cases but was reversible in all and was treatment limiting in one patient only. The 100-day mortality and 1-year mortality after initiation of OPAT were 0% and 6%, respectively. In a selected population, and after confirmation of initial response to treatment, our data support the use of OPAT with LAmB for treatment of IFI in an intermittent dosing regimen.
Collapse
Affiliation(s)
- Robert J van de Peppel
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands.,Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Alexander Schauwvlieghe
- Department of Internal Medicine, Section of Infectious Diseases, Erasmus MC, University Medical Center Rotterdam
| | - Ruth Van Daele
- Pharmacy Department, University Hospitals Leuven and Department of Pharmaceutical and Pharmacological Sciences, Clinical Pharmacology and Pharmacotherapy, KU Leuven, Belgium
| | - Isabel Spriet
- Pharmacy Department, University Hospitals Leuven and Department of Pharmaceutical and Pharmacological Sciences, Clinical Pharmacology and Pharmacotherapy, KU Leuven, Belgium
| | - Jan W Van't Wout
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Roger J Brüggemann
- Department of Pharmacy and Radboud Institute for Health Sciences, Radboud University Medical Center; Center of Expertise in Mycology Radboud / CWZ, Radboud University Medical Center Nijmegen, The Netherlands
| | - Bart J A Rijnders
- Department of Internal Medicine, Section of Infectious Diseases, Erasmus MC, University Medical Center Rotterdam
| | - Bart J C Hendriks
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center
| | - Mark G J de Boer
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
15
|
Abstract
Neonates and immunosuppressed/immunocompromised pediatric patients are at high risk of invasive fungal diseases. Appropriate antifungal selection and optimized dosing are imperative to the successful prevention and treatment of these life-threatening infections. Conventional amphotericin B was the mainstay of antifungal therapy for many decades, but dose-limiting nephrotoxicity and infusion-related adverse events impeded its use. Despite the development of several new antifungal classes and agents in the past 20 years, and their now routine use in at-risk pediatric populations, data to guide the optimal dosing of antifungals in children are limited. This paper reviews the spectra of activity for approved antifungal agents and summarizes the current literature specific to pediatric patients regarding pharmacokinetic/pharmacodynamic data, dosing, and therapeutic drug monitoring.
Collapse
Affiliation(s)
- Kevin J Downes
- Division of Infectious Diseases, Children's Hospital of Philadelphia, 2716 South Street, Suite 10360, Philadelphia, PA, 19146, USA.
- Center for Pediatric Clinical Effectiveness, Children's Hospital of Philadelphia, Philadelphia, PA, USA.
- Center for Clinical Pharmacology, Children's Hospital of Philadelphia, Philadelphia, PA, USA.
- Department of Pediatrics, Perelman School of Medicine of the University of Pennsylvania, Philadelphia, PA, USA.
| | - Brian T Fisher
- Division of Infectious Diseases, Children's Hospital of Philadelphia, 2716 South Street, Suite 10360, Philadelphia, PA, 19146, USA
- Center for Pediatric Clinical Effectiveness, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pediatrics, Perelman School of Medicine of the University of Pennsylvania, Philadelphia, PA, USA
| | - Nicole R Zane
- Center for Clinical Pharmacology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| |
Collapse
|
16
|
Stevens VM, Mueller SW, Reynolds PM, MacLaren R, Kiser TH. Extrapolating Antifungal Animal Data to Humans - Is it reliable? CURRENT FUNGAL INFECTION REPORTS 2020; 14:50-62. [PMID: 32201545 PMCID: PMC7083583 DOI: 10.1007/s12281-020-00370-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
PURPOSE OF REVIEW This article aimed to review animal models of antifungals and identifies human literature to assess if the extrapolation of results is reliable. RECENT FINDINGS Animal studies have helped identify AUC/MIC targets for new drugs and formulations such as isavuconazole and delayed release posaconazole that have translated to successful outcomes in humans. Models have also been influential in the identification of possible combination therapies for the treatment of aspergillosis, such as voriconazole and echinocandins. However, challenges are endured with animal models when it comes to replicating the pharmacokinetics of humans which has been exemplified with the newest itraconazole formulation. Additionally, animal models have displayed a survival benefit with the use of iron chelators and amphotericin for mucormycosis which was not demonstrated in humans. SUMMARY Animal models have been a staple in the development and optimization of antifungal agents. They afford the ability to investigate uncommon diseases, such as invasive fungal infections, that would otherwise take years and many resources to complete. Although there are many benefits of animal models there are also shortcomings. This is why the reliability of extrapolating data from animal models to humans is often scrutinized.
Collapse
Affiliation(s)
- Victoria M Stevens
- Department of Clinical Pharmacy, University of Colorado Skaggs School of Pharmacy and Pharmaceutical Sciences, 12850 East Montview Boulevard, Mail Stop C238, Aurora, CO 80045, USA
| | - Scott W Mueller
- Department of Clinical Pharmacy, University of Colorado Skaggs School of Pharmacy and Pharmaceutical Sciences, 12850 East Montview Boulevard, Mail Stop C238, Aurora, CO 80045, USA
| | - Paul M Reynolds
- Department of Clinical Pharmacy, University of Colorado Skaggs School of Pharmacy and Pharmaceutical Sciences, 12850 East Montview Boulevard, Mail Stop C238, Aurora, CO 80045, USA
| | - Robert MacLaren
- Department of Clinical Pharmacy, University of Colorado Skaggs School of Pharmacy and Pharmaceutical Sciences, 12850 East Montview Boulevard, Mail Stop C238, Aurora, CO 80045, USA
| | - Tyree H Kiser
- Department of Clinical Pharmacy, University of Colorado Skaggs School of Pharmacy and Pharmaceutical Sciences, 12850 East Montview Boulevard, Mail Stop C238, Aurora, CO 80045, USA
| |
Collapse
|
17
|
Bochennek K, Luckowitsch M, Lehrnbecher T. Recent advances and future directions in the management of the immunocompromised host. Semin Oncol 2020; 47:40-47. [DOI: 10.1053/j.seminoncol.2020.02.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 02/05/2020] [Accepted: 02/05/2020] [Indexed: 02/07/2023]
|
18
|
Lehrnbecher T. The clinical management of invasive mold infection in children with cancer or undergoing hematopoietic stem cell transplantation. Expert Rev Anti Infect Ther 2019; 17:489-499. [DOI: 10.1080/14787210.2019.1626718] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Thomas Lehrnbecher
- Division of Pediatric Hematology and Oncology, Hospital for Children and Adolescents, Johann Wolfgang Goethe-University, Frankfurt, Germany
| |
Collapse
|