Skip to main content
Erschienen in: Italian Journal of Pediatrics 1/2023

Open Access 01.12.2023 | Review

Management of pulmonary aspergillosis in children: a systematic review

verfasst von: Vito Terlizzi, Marco Antonio Motisi, Roberta Pellegrino, Luisa Galli, Giovanni Taccetti, Elena Chiappini

Erschienen in: Italian Journal of Pediatrics | Ausgabe 1/2023

Abstract

Invasive pulmonary aspergillosis (IPA) is a severe condition in immunocompromised children, but the optimal management is still under debate. In order to better clarify this issue, a literature search was performed through MEDLINE/PubMed database to describe current risk factors and diagnostic, therapeutic and prophylactic tools for invasive pulmonary aspergillosis (IPA) in the paediatric age. Observational studies and clinical trials regarding diagnosis, treatment and prophylaxis were considered, and results were summarised. Five clinical trials and 25 observational studies (4453 patients) were included.
Haematological malignancies, previous organ transplant and other primary or acquired immunodeficiency were identified as risk factors for IPA in children.
Current diagnostic criteria distinguish between "proven", "probable" and "possible" disease. Consecutive galactomannan assays have good sensitivity and specificity, especially when performed on broncho-alveolar lavage. At the same time, β-D-glucan should not be used since cut-off in children is unclear. PCR assays cannot currently be recommended for routine use.
Voriconazole is the recommended first-line agent for IPA in children older than 2 years of age. Liposomal amphotericin B is preferred in younger patients or cases of intolerance to voriconazole. Its plasma concentrations should be monitored throughout the treatment. The optimal duration of therapy has yet to be determined. Posaconazole is the preferred prophylactic agent in children older than 13 years old, whereas oral voriconazole or itraconazole are the drugs of choice for those between 2–12 years. Further good-quality studies are warranted to improve clinical practice.
Hinweise

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
IPA
Invasive pulmonary aspergillosis
PCR
Polymerase chain reaction
RCT
Randomized controlled trials
CF
Cystic fibrosis
PRISMA
Preferred reporting items for systematic reviews and metanalyses
AML
Acute myeloid leukaemia
HSCT
Hematopoietic stem cells transplantation
GVHD
Graft versus host disease
OR
Odds ratio
CI
Confidence interval
CGD
Chronic granulomatous disease
GM
Galactomannan antigen
BAL
Bronchoalveolar lavage
PPV
Positive predictive value
NPV
Negative predictive value
CT
Computed tomography
DNA
Deoxyribonucleic acid

Main text

Introduction

Aspergillus spp. is a ubiquitous, slow-growing mold that commonly colonises the respiratory tract. Depending on the host immune status and lung structure, it can manifest as different clinical entities, such as aspergilloma, allergic bronchopulmonary aspergillosis (ABPA), allergic sinusitis, invasive aspergillosis, chronic pulmonary aspergillosis [1]. Allergic sinusitis and ABPA are allergic responses to colonisation by Aspergillus spp. occurring in immunocompetent patients. In particular, ABPA affects children with asthma, causing frequent flare-ups, and/or cystic fibrosis (CF) [2]. Invasive pulmonary aspergillosis occurs in immunocompromised children with impaired neutrophil and T-lymphocyte function and children with chronic pulmonary diseases or CF. This severe disease requires prompt treatment, but the diagnosis and management of IPA in children are still challenging and often delayed. Environmental and medical prophylaxis is essential in patients with risk factors, and in such cases, clinical and radiological suspicion is sufficient to initiate empirical therapy with antifungal agents. Since most of the available studies concerning IPA are carried out in adults, we performed a systematic literature review with the aim of summarising the currently available data regarding IPA in pediatric age with a focus on diagnostic tools, treatment and prophylaxis.

Methods

The authors identified the following five key questions:
1.
What are the main risk factors for IPA in children?
 
2.
What are the main diagnostic tools for IPA in children?
 
3.
What is the first-line agent for IPA in children?
 
4.
What is the optimal duration of treatment for IPA in children?
 
5.
What are the main prophylactic drugs used for IPA in children?
 
A systematic review of the literature was performed in line with the Preferred Reporting Items for Systematic Reviews and Meta-analyses (PRISMA) guideline recommendations [3]. The research was conducted through MEDLINE/PubMed, including articles published from the 1st of January 2002 to the 21st of December 2021. References of all relevant articles were also evaluated, and pertinent articles were included. The search strings were as follows: “(Invasive[Title/Abstract]) AND (Aspergillosis[Title/Abstract]) AND (Children[Title/Abstract] OR Paediatric[Title/Abstract])” and “(Invasive[Title/Abstract]) AND (Aspergillosis[Title/Abstract]) AND (Pulmonary[Title/Abstract] OR Lung [Title/Abstract]) AND (Children[Title/Abstract] OR Paediatric[Title/Abstract])”.

Inclusion and exclusion criteria

The research was restricted to English language. Articles reporting risk factors, diagnostic tools, treatment and prophylaxis for IPA in paediatric population (age < 18 years) with a sample size greater than ten were included independently from the study design. Review articles, case reports, commentaries, editorials, letters to the author and pre-print records were excluded as well as studies referring to adult populations.

Data extraction

Duplicate publications were removed, then two authors separately (RP and MAM) checked the titles and abstracts and removed irrelevant studies according to the inclusion and exclusion criteria. Pertinent articles from the bibliographic references of the selected studies were also considered, and an additional review of the literature was performed prior to final drafting. Articles were categorized according to the study design. Data about IPA risk factors, diagnostic tools, treatment, and prophylaxis were extracted.

Quality assessment

The Jadad scale was used to assess quality for RCTs [4], while the Methodological Index for Non-randomized Studies (MINORS) was used for non-randomized ones [5]. Observational studies were evaluated for adherence to Strengthening the Reporting of Observational Studies in Epidemiology (STROBE) recommendations [6].

Results

Five clinical trial (4 RCT and 1 non-randomized trial) and 25 observational studies were included in the review (Fig. 1). The quality assessment of selected studies is reported in Figs. 2 and 3. The characteristics and findings of selected studies are summarized in Table 1.
Table 1
Summary of findings
Author
Year
Country
Aim
Study design
Number of patients
Age
Results
Anantasit et al. [7]
2018
Thailand
To validate EORT/MSG 2008 definition in paediatric population
Histology vs EORTC/MSG 2008
Retrospective Cross-sectional
256
1 m- 18 y
EORT/MSG 2008 sensitivity 100% specificity 36%
PPV 33%
NPV 100%
EORT/MSG useful as screening tool
Neutropenia described as risk factor for IPA
Avcu et al. [8]
2017
Turkey
To determine the utility of serum GM monitoring in early diagnosis of IA and its role in the management of children with ALL
Retrospective cohort study
141
3264 samples
Median age 55 m (range 3–208 m)
False positive: 52.1%
Multiple consecutive positive tests increased the incidence of true-positive tests and introduction of antifungal therapy
Badiee et al. [9]
2012
Iran
To evaluate the diagnostic potential of EIA for GM, nested PCR and BDG test
Prospective matched cohort study
62 patients
230 samples
36 pts (129 samples) with IA suspicion
26 pts (101 samples) controls
Mean age 9.3 y
Galactomannan EIA: sensitivity 90%, specificity 92%, PPV 81.8% NPV 96%, likelihood ratios for positive results 11.25, for negative results 0.1
beta–D–glucan: sensitivity 50%, specificity 46%, PPV 26%, NPV 70.6%, likelihood ratios for positive results 0.9, for negative results 0.9
nested-PCR: sensitivity 80%, specificity 96.2%, PPV 88.9%, NPV 92.6%, likelihood ratios for positive results 21, and negative results 0.2
Galactomannan and nested-PCR tests are useful as non-invasive methods for diagnosis of IA in children. Beta–D–glucan test is not an efficient diagnostic tool in those with hematologic disorders
Burgos et al. [10]
2008
US
To describe risk factors, diagnostic tools, treatments and outcomes of IA in children
Retrospective cross-sectional study
139 pts
Median age 10.1 y (17d-18y)
A. fumigatus was the most often reported species
Risk factors: immunosuppressive therapies and allogenic HSCT
Most common site: lungs (59%)
Most frequent radiologic finding: nodules 34.6% with 2.2% showing the crescent sign, 11% the halo sign and 43.1% cavitation
Treatment: 45.8% received more than 3 concomitant antifungal agents, no superiority among antifungal agents was found
Caglar et al. [11]
2020
Turkey
To evaluate the diagnostic value of serum GM positivity for IA in children
Retrospective cohort study
70 patients
104 samples
Median age 5 y (1–16)
Consecutive GM positivity has higher PPVs independently from the cut-off value chosen
Choi et al. [12]
2013
Korea
To investigate the use of GM antigen assay as diagnostic tool in pediatric cancer and HCT patients; to assess the characteristics of patients with IA
Retrospective controlled cohort study
83 patients
23 IA group 60 non-IA group
640 samples
Median age
IA 12.3 y (0.7–18.4)
Non-IA 6.4y (0.3–18.7)
The false-positive rate was 18.3%
Being younger than 3 years of age, having a solid tumor, and receiving HCT within 4 weeks from the test caused false-positive results (p < 0.05)
The most common clinical site of IA was the lung (91.3%), and consolidation was the most frequent finding in chest CT scans (36.8%). The mortality at 12 weeks was 43.5%
Having a positive GM assay at least twice is useful in diagnosing IA in pediatric patients with cancer and HCT recipients
de Mol et al. [13
2013
Netherlands
To study the diagnostic value of BAL GM in immunocompromised children
Retrospective cross sectional study
47 pts
47 bronchoscopies
Median age 9.8 (1.1–18.2) y
BAL GM for proven and probable IPA:
Sensitivity 82.4%, specificity 87.5%, PPV 82.4, NPV 87.5%
A significant relation for BAL GM and abnormal chest CT (p = 0.01)
BAL GM and serum GM correlated significantly
BAL GM test had good diagnostic value in children with suspected IPA. The decision to continue or start antifungal therapy was mainly determined by the clinical suspicion of IPA based on chest CT-outcome, serum GM index values and failure of antibiotic therapy
Dinand et al. [14]
2016
India
To evaluate the use and optimal serum GM cut-off in children
Prospective cohort study
145 pts
211 febrile episodes
Median age 5 (0.5–19) y
Serum GA is sensitive to diagnose IA in pediatric patients with excellent NPV with a cut-off of 0.7. Two consecutive values of 0.7 increases specificity to 91.0%
Doring et al. [15]
2012
Germany
To analyse safety and efficacy of CAS and L-AmB in HSCT patients
Retrospective matched cohort study
60 pts received CAS
60 pts received L-AmB
Median age
Cas group 9.5y
L-AmB group 7.5y
Similar efficacy between prophylaxis with CAS and L-AmB after allogenic HSCT
More drug-related side effects and an increased need for oral supplementation with potassium, sodium bicarbonate and calcium upon discharge in L-AmB receiving group
Doring et al. [16]
2015
Germany
To assess safety, feasibility, and
efficacy of posaconazole compared to fluconazole and itraconazole in neutropenic children and adolescent
Retrospective matched cohort study
93 pts
31 fluconazole
32 itraconazole
30 posaconazole
Median age 12y (9 m-17.7y)
Posaconazole, fluconazole, and itraconazole are comparably effective in preventing invasive fungal infections in children
Larger studies are required to define dose recommendations
No statistical significant differences found in adverse events
Fisher et al. [17]
2012
US
To evaluate GM EIA as diagnostic tool in children after intensive chemotherapy or HSCT
Multicentre prospective cohort study
213 patients
1865 serum samples from 198 pts
886 urine samples from 183 pts
7 BAL samples from 4 pts
7.8 y
Serum GM testing specificity 95%
Urine GM testing specificity 80%
The urine test resulted in a higher false positivity rate, but it successfully identified the only case of probable IA
Screening for GM, or a related antigen in urine, needs to be further evaluated as it may be useful in surveillance strategies
Fisher et al. [18]
2019
US and Canada
To compare the efficacy of CAS vs. fluconazole prophylaxis against proven or probable invasive fungal disease and IA in neutropenic patients following AML chemotherapy
Multicentre randomized open label clinical trial
257 CAS
260 fluconazole
Median age 9 (0–36) y
Prophylaxis with CAS compared with fluconazole resulted in significantly lower incidence of invasive fungal disease and proven and probable IA
Fisher et al. [19]
2021
US
To assess surveillance testing with GM EIA and BDG assay in children with AML receiving antifungal prophylaxis
Prospective cohort study
425 pts
209 fluconazole
216 CAS
6103 samples
Median age 10(0–25) y
NPV > 99% for GM EIA and BDG test alone and in combination
Sensitivity and PPV 0%
GM EIA and BDG test should be discouraged for surveillance in patients with AML receiving antifungal prophylaxis
Gefen et al. [20]
2015
Israel
To investigate serial serum GM assay screening on IPA diagnosis in children with HSCT or high risk leukemia
Prospective cohort study
34 pts
510 samples in neutropenic children
Median age 8.5 y (6 m-19y)
GM assay: sensitivity 0.8, specificity 0.66, PPV 0.22 and NPV 0.96
Han et al. [21]
2015
Korea
To characterize IPA in children with hematological/oncological disorders
Retrospective matched cohort study
166 pts
 
Neutropenia lasting more than 2 weeks (51.4% vs. 21.9%, p < 0.001) and halo signs at chest CT (78.4% vs. 40.7%, p < 0.001) were more frequent among children with IPA
Early use of chest CT in children at risk of prolonged neutropenia could be helpful for early IPA diagnosis
Herbrecht et al. [22]
2002
Multicentre
Invasive Fungal Infection Group of EORTC
To compare voriconazole with AmB as primary therapy of IPA
Randomized, unblinded clinical trial
144 pts voriconazole
133 AmB
Mean age voriconazole group: 48.5 (13–79)y
AmB group: 50.5 (12–75) y
Voriconazole led to better responses, improving survival with less severe side effects than amphotericin B
Itsaradisaikul et al. [23]
2021
Thailand
To evaluate 1-year incidence of IFD after itraconazole prophylaxis in HSCT children; to identify risk factors, etiology and adverse events
Retrospective cohort study
170 pts
Median age 8.43 (5.41–12.36) y
Itraconazole did not showed a excellent efficacy in preventing IFD after HSCT. It could be used in resource-limited settings. It requires appropriate drug level monitoring if used
Jha et al. [24]
2013
India
To evaluate the role of GM assay in IA diagnosis in children on treatment for hematological malignancies and to identify the best cut-off values
Prospective cohort study
78 pts
100 ferbile episodes
Mean age 6.1 y (1.5–13)
Best results with cut-off value of 1.0
GM assay (cut-off value 1.0):
Sensitivity 60%, specificity 93%, PPV 75, NPV 87
A higher value of GM related with pulmonary nodules (p = 0.037) and mortality (p = 0.001)
Kato et al. [25]
2016
Japan
To identify the daily therapeutic dose in children; to analyze association between voriconazole concentration and clinical outcomes
Retrospective cohort study
20 pts
111 samples
Median age 9.5 (0–17) y
younger age and oral administration were
associated with lower plasma voriconazole concentrations (p < 0.01). Unfavourable
outcome was associated with low concentrations of voriconazole (p = 0.01)
Higher doses are required in younger children and in case of oral administration
Kazakou et al. [26]
2020
Greece
To evaluate the incidence of IFD in children with hematological malignancies and determine the clinical characteristics, risk factors, diagnosis, treatment efficacy and outcome
Retrospective cohort study
297 pts
Mean age 6.64 (2–13) y
Most common underlying disease: ALL (79%)
Most common site of infection: lungs (66.7%)
Identified species: Aspergillus spp. (58.3%)
Most prescribed treatment: L-AmB
The crude mortality rate was 33.3%
Lee et al. [27]
2017
Korea
To determine safety and efficacy of the combination of Voriconazole and CAS to treat IFDs
Retrospective cross-sectional study
22 pts
Mean age 5
3 (0.8–13.3) y
Voriconazole plus CAS is an effective and safe treatment for serious IFD in children with leukemia
Loeffler et al. [28]
2017
Germany
To determine the use of GM assay combined with PCR assay in HSCT recipients
Retrospective cohort study
39 pts
543 samples
Median age
male: 9.5 (4–21) y
female: 10 (3–19) y
GM assay: specificity 89%, sensitivity 67%, PPV 50% NPV 100%
PCR assay: specificity 63%%, sensitivity 100%, PPV 27% NPV 100%
combined monitoring for GM and fungal DNA results in a higher diagnostic accuracy
Maertens et al. [29]
2010
US and Europe
To evaluate CAS vs L-AmB for Empiric antifungal therapy in children with persistent fever and neutropenia
Randomized double blind clinical trial
83 pts
57 CAS
26 L-AmB
Range 2–17 y
CAS and L-AmB were comparable in tolerability, safety and efficacy as empiric antimicotic therapy
Maron et al. [30]
2013
US
To compare etiology, predisposing factors and outcomes of IFD in AML patients before and after implementation of voriconazole prophylaxis
Retrospective cohort study
19 pts
AML97 (no fungal prophylaxis) = 12 pts
AML02 (voriconazole prophylaxis) = 11 pts
Median age
AML97: 11 (0.3–21) y
AML02: 8(1–19) y
Voriconazole prophylaxis was associated with improved survival and a significant reduction in aspergillosis
Martin et al. [31]
2017
Multicentre Asia, Europe and North America
To evaluate safety, efficacy and exposure–response of voriconazole as treatment for IA, IC and EC
Prospective open-label, non-comparative phase 3 study
53 pts
31 IA
22 IC/EC
Mean age
IA 11.9 (SD 3.5)
IC/EC 9.5 (SD 4.5)
In IA cohort: 22.6% treatment related hepatic AE and 16.1% visual AE; all-causality mortality 14.3% at week 6, no deaths attributed to voriconazole
Voriconazole is effective in patients with IA with a favourable risk–benefit balance
Mohammadi et al. [32]
2015
Iran
To evaluate the efficacy of BAL GM in immunocompromised patients
prospective case–control study
16 pts immunocompromised, with possible/proven IPA by EORT/MSG criteria
54 controls
Mean age (IPA pts): 8.4 (11–15) y
BAL GM using an OD index of ≥ 0.5:
Sensitivity 87.5% PPV 93.33%
High diagnostic value of BAL GM in immunocompromised children with IPA
Qiu et al. [33]
2019
China
To evaluate the diagnostic value for IPA of serum GM combined with CT in children after HSCT
Retrospective case control study
46 cases
95 controls
Mean age
Cases 7 ± 3.7 y
Controls 6.2 ± 3.5y
GM testing combined with CT evaluation: PPV of 0.764, and NPV of 0.872, Sensitivity 0.793, and specificity 0.852
The combination of serum GM and chest CT might be used for early diagnosis of IPA in HSCT patients
Vrioni et al. [34]
2018
Greece
To define the use of GM serum assay and PCR as routine methods for IA in immunosuppressed children
Prospective cohort study
156 pts
744 samples
Age range 5 m-14y
Agreement of the two methods: 90% of pts, 96.1% of samples
The combination of GM and PCR had a high diagnostic accuracy in consecutive samples (twice weekly)
Zaoutis et al. [35]
2006
US
To describe the incidence and outcomes of IA in children
Retrospective cohort study
666 pts
Median age 13 (IQR 8–15)
Highest incidence of IA in children with HSCT (4.5%) and AML (4%). The overall in- mortality of immunocompromised children was 18%. Children with malignancy and IA were at higher risk for death
Children with IA had a longer hospital stay and higher hospital charges
Abbreviations: ALL Acute Lymphoblastic Leukemia, AML Acute Myeloid Leukemia, AML97 and AML02: multicentre protocols for pediatric patients with AML, BAL Bronchoalveolar lavage, BDG β-D-glucan, CAS Caspofungin, CT Computed tomography, d days, EIA Ezyme Immunoassay, EC Esophageal candidiasis, EORTC/MSG European Organization for Research and Treatment of Cancer/Invasive Fungal Infections Cooperative Group and The National Institute of Allergy and Infectious Disease Mycoses Study Group, GM galactomannan, HSCT Hematopoietic Stem Cell Transplant, IA Invasive Aspergillosis, IC Invasive candidiasis, IFD Invasive Fungal Disease, IQR Interquartile range, L-AmB Liposomal amphotericin B, m months, NPV Negative predictive value, OD Optical density, PCR Polymerase Chain Reaction, PPV Predictive positive value, Pts Patients, y years, US United States

What are the main risk factors for IPA in children?

In immune-competent hosts, occasional colonisation of fungal conidia is controlled by the immune system. In contrast, in immune-compromised subjects, an invasive infection can occur, primarily affecting the lungs. However, dissemination to the central nervous system is reported in up to 30% of cases. The IPA development depends on the patient's immune status and lung parenchyma characteristics (Table 2).
Table 2
Host factors for invasive pulmonary aspergillosis
Host factors for invasive pulmonary aspergillosis
1) Prolonged neutropenia (< 500 cells/mm3 for > 10 days)
2) Transplantation (higher risk for lung and hematopoietic stem cell transplantation)
3) Prolonged (> 3 weeks) high-dose corticosteroid therapy in the past 60 days
4) Treatment with other T-cell immunosuppressants
5) Treatment with B-cell immunosuppressants
6) Hematological malignancy (higher risk for leukemia)
7) Severe primary immunodeficiencies (e.g., CGD, WAS)
8) Acute grade III-IV GVHD with gut, lung or liver involvement and steroid-resistant
Abbreviations: CGD Chronic granulomatous disease, WAS Wiskott-Aldrich syndrome, GVHD Graft-versus-host disease
IPA represents an emerging problem and is one of the leading causes of morbidity and mortality in immune-compromised patients. The incidence of IPA in children receiving chemotherapy is high. It is associated with increased morbidity and death, with highest rates in patients with acute myeloid leukaemia (AML), recurrent leukaemia, and those undergoing hematopoietic stem cell transplantation (HSCT) [10, 21, 26]. A large multicenter study conducted by Zaoutis et al. in 2006 including 666 children with invasive aspergillosis found that 60–75% of cases are oncologic patients with mortality rates as high as 85% [35]. The risk was higher in case of allogeneic transplantation than in autologous transplantation and in cases of severe graft versus host disease (GVHD). One of the larger cohorts of patients with IPA was described by Burgos et al. in a multicenter retrospective analysis in 2008; they examined 139 paediatric patients with invasive aspergillosis, 80% of which had lung involvement [10]. The most common underlying conditions were haematological malignancies (87/139, 62.6%), followed by inherited immunodeficiencies (16/139, 11.5%), solid organ transplant (16/139, 11.5%), solid tumour (9/139, 6.5%). In particular, 51 patients (26.6%) underwent allogenic HSCT, which was identified as the most critical risk factor for overall mortality in invasive aspergillosis (OR 6.14 – 95% CI 2.67–16.21). Severe neutropenia, defined as neutrophil count below 500 cells/mm3, is the main single risk factor for the development of IPA; in the abovementioned study, it accounted for about 59% of aspergillosis cases. In addition, most hemato-oncology patients presented additional immunosuppression due to prolonged therapy with high-dose steroids or immunosuppressive drugs such as cyclosporine or tacrolimus [10, 26].
Considering primary immunodeficiencies, invasive aspergillosis may be the presenting manifestation or a frequent complication in the first two decades of life in children with chronic granulomatous disease (CDG), a condition caused by neutrophil dysfunction. In such patients, Aspergillus spp. can cause brain abscesses, osteomyelitis and lung damage. Invasive aspergillosis may also occur in children with Wiskott-Aldrich syndrome (WAS), in which neutrophil chemotaxis and lymphocyte function is impaired. Burgos et al. found that CDG accounted for almost 50% of invasive aspergillosis cases among children with primary immunodeficiency (7/16). In contrast WAS was found to be the predominant underlying condition (81/122, 66%) in the larger study by Zaoutis et al., followed by CDG (21/122, 17%) [10, 35]. Among all children with invasive aspergillosis, the highest incidence was observed in those with WAS (30%), followed by CGD (6.5%), allogenic HSCT (4.5%) and AML (3.7%) [35]. Nevertheless, immunodeficiency is not the only risk factor for IPA, since patients hospitalised in intensive care units, children with chronic obstructive pulmonary disease, emphysema, or those affected by chronic diseases such as CF, are also at increased risk. Nevertheless, Zaoutis et al. observed that only 5 out of over 11,000 patients with CF were diagnosed with IPA [35].

What are the main diagnostic tools for IPA in children?

The diagnosis of IPA still represents a challenge for the clinician today. Despite the introduction of new diagnostic techniques, the high mortality rate of this condition is mainly due to diagnostic delay [12]. An early diagnosis and prompt therapy is crucial for a better outcome, especially in the immune-compromised child. Maintaining a high index of suspicion in patients with multiple risk factors is essential. An in-depth diagnostic investigation is necessary in case of fever unresponsive to antibiotic therapy, or cough with sputum and dyspnoea in high-risk children. Chest pain with pleural involvement (due to small pulmonary infarcts) and haemoptysis may be associated with the clinical picture of IPA. When infection disseminates to the central nervous system, seizures or radiological alterations consistent with cerebral infarcts, intracranial haemorrhages, or epidural abscesses may occur.
No specific biochemical and/or instrumental tests allow a diagnosis of certainty. Therefore, diagnostic tests should be performed sequentially starting with the least invasive ones. The result of each test should be considered according to the clinical and immune status of the patient. Currently validated diagnostic criteria for adults, which are also used in some paediatric studies [12, 13] distinguish between “proven”, “probable” and “possible” IPA. In a recent Consensus of the European Organization for Research and Treatment of Cancer and the Mycoses Study Group (EORTC/MSG), the definition of invasive fungal disease has been revised, with no change from the previous 2008 classification (Table 3) [36]. These criteria have shown to have an excellent sensitivity but low specificity in detecting possible or probable cases of IPA [7]. The least invasive test for identification of aspergillus infection is the blood assay of galactomannan antigen (GM), a wall component released in the blood due to its growth. GM can also be assayed in other biological fluids, such as bronchoalveolar lavage fluid (BAL) or cerebrospinal fluid, in rare cases of neurological involvement, being more reliable than the blood value [12, 13]. The sensitivity and specificity of the blood assay depend on various components, such as the underlying pathology, current therapies or cut-off used by the laboratory, since there is currently no consensus about negative values. Choi et al. analysed 749 blood samples from 99 oncological children and showed that blood assay of GM was more reliable in cancer patients in detecting invasive aspergillosis (sensitivity: 91.3%; specificity: 81.7%; false positives: 18%) [12]. Similar results were found in the studies of Badiee et al. and Fisher et al. on 62 and 198 paediatric haemato-oncology patients, respectively [9, 17]. Recent studies showed variable sensitivity, specificity and positive predictive value (PPV) of single serum GM determination. On the other hand, consecutive positive tests had higher PPV, especially in a compatible clinical and radiological context [8, 11, 20, 24]. This variability is, to some extent, linked to that of the cut-off used to define GM positivity. In the prospective study of Dinand et al. on 145 neutropenic patients, the optimal cut-off value for single GM determination was found to be 0.7 with sensitivity and specificity both around 82% and negative predictive value (NPV) of 94%, and specificity increased to 91% in case of positivity on a consecutive test [14]. In a recent cross-sectional study of Çağlar et al. on 70 patients with haematological malignancies, consecutive GM positivity displayed higher PPVs independently from the cut-off value chosen [11]. Considering GM on BAL, Mohammadi et al. reported sensitivity and positive predictive values of 87.5% and 93.33% respectively, using a cut-off of ≥ 0.5. Moreover in 7 out of 16 cases of IPA, serum GM was negative, while their BAL GM was positive [32].
Table 3
Diagnostic criteria for invasive aspergillosis [10]
Diagnosis
Criteria
Certain
Histological or cytological evaluation of lung tissue with hyphae on needle aspiration or biopsy in which hyphae or melanized yeast-like forms are associated to tissue damage
or
Positive culture test for Aspergillus on pulmonary specimen taken by sterile procedure
and
Clinical or radiologic abnormalities consistent with infection
Probable
At least 1 host factor (tab. 2)
and
Mycological evidence
- positive microscopy or culture for Aspergillus on sputum, BAL bronchial brush, or aspirate
- positive Aspergillus PCR (at least 2 tests)
- positive antigenic assaya
AND
Clinical criteria consistent with infectionb
Possible
At least 1 host factor (tab.2)
and
Clinical criteria compatible with infectionb
aPositive antigenic assay: detection of galactomannan in plasma, serum, BAL, or CSF. β-D-glucan was not considered to provide mycological evidence of any invasive mold disease
bClinical criteria compatible with infection: characteristic infiltrates on CT (dense, well-circumscribed lesions with or without halo sign, air crescent sign, or cavity), tracheobronchitis diagnosed by bronchoscopy, or infiltrates that are uncharacteristic but associated with specific pulmonary symptoms or signs (e.g., pleural pain, haemoptysis)
Abbreviations: BAL Bronchoalveolar lavage, CSF Cerebrospinal fluid, CT Computed tomography, PCR polymerase chain reaction
Another Aspergillus antigen is β-D-glucan, which is shared with other fungal species such as Candida spp. and Pneumocystis spp. Data available in children are scarce, and an optimal cut-off is unknown, as mean β-D-glucan levels are higher in immunocompetent children than in adults. The combined assay of the two wall components may the reliability of the tests [9] even if, to date, β-D-glucan is not recommended for screening or evaluating suspected IPA in high-risk patients. At last, neither GM nor β-D-glucan (alone or in combination) can be used as a screening marker in neutropenic patients undergoing antifungal prophylaxis, since none of them has shown an acceptable sensitivity [19].
Evidence regarding the use of other biochemical parameters, such as total IgE, Aspergillus-specific IgE or peripheral eosinophil counts as significant support for the diagnosis of IPA is lacking.
Radiological examinations are essential for the diagnosis of IPA. Nevertheless, recent studies showed that the current radiological criteria used in adults are not applicable in children [10, 12]: radiographic findings considered typical of IPA in adults are not seen in the majority of children with IPA, and unspecific findings are detected more often in immunocompromised children. I In case of multiple nonspecific nodules t chest-xrays (up to 35% of cases) [10], a differential diagnosis with viral (from Cytomegalovirus or Adenovirus) or bacterial (Nocardia) pneumonia is required. Similarly, high-resolution chest computed tomography (HRCT), the most useful imaging tool, rarely evidences pathognomonic lesions of IPA in children, such as “halo sign” (area of ground-glass opacity surrounding a nodule), “air crescent sign” (area of increasing radiolucency in a region of nodular opacity), or cavitary lesions [10, 12]. A single-centre case–control study including 141 children with neutropenic fever lasting more than 96 h showed that the combination use of chest CT scan and serum GM testing was useful for early diagnosis, with PPV and NPV of 76% and 87%, respectively [33]. In the multicenter analysis of Burgos et al., including 110 children with IPA, 61 of them had 2 or more radiological findings on chest CT or plain radiograph, with pulmonary nodules being the most common; (65/110, 59%); on the other hand, halo sign and air crescent were found in smaller percentages of patients (10.9% and 2.2% respectively) [10]. Thus, radiological findings are often unsatisfactory and further examination is required for diagnostic confirmation. Molecular testing through Polymerase Chain Reaction (PCR) on blood or BAL has recently been included among diagnostic criteria for probable aspergillosis. However, its role in patient management has not been established yet. Badiee et al. reported a high NPV of Aspergillus-specific PCR as a screening tool [9], in line with two other studies showing that a combination of GM and PCR testing could be used for screening or diagnostic purposes, especially when tested on consecutive samples [28, 34], However, despite promising results, Aspergillus PCR is not recommended for routine use yet, since its standardization and validation are lacking.
The current gold standard for the diagnosis of IPA is lung biopsy. It is an invasive exam difficult to perform in clinically compromised patients, considering the frequent association between thrombocytopenia and advanced forms of IPA. Therefore, lung biopsy should be performed only when less invasive procedures have not been conclusive. In most cases, lung biopsy is performed through a CT-guided transbronchial route with BAL collection. Since the collected sample is often quantitatively insufficient and the sensitivity of culture examinations is poor, PCR techniques have been introduced to identify Aspergillus DNA on histological samples [9].
Lastly, hyphae in lung tissue or a positive culture for Aspergillus spp. on the same site or on normally sterile biological fluids allows the diagnosis of IPA in patients with a suggestive clinical or radiological picture. In high-risk children with a clinical and radiological suspect of IPA, GM antigen positivity on serum or BAL or fungal growth in BAL are required for IPA diagnosis and in these cases lung biopsy is unnecessary.

What is the first-line agent for IPA in children?

The mortality rate of IPA in untreated paediatric patients is close to 100% and remains very high even with aggressive drug therapy. For this reason, empirically based antifungal therapy must be started as soon as possible in children at high risk of developing invasive forms of aspergillosis or otherwise undergoing diagnostic evaluation [7, 27]. In paediatric age, the major difficulties in the therapeutic choice are related to the lack of RCTs on large sample sizes and the paucity of unambiguous data regarding the first-choice drug class, the duration of therapy, and the outcomes to evaluate the clinical response. Most of the available data refer to studies carried out on adults. To date, the first-choice treatment options in children older than 2 years of age are voriconazole, and liposomal amphotericin B. Voriconazole is not approved in children younger than 2 years of age, and the optimal dose is unclear, hence liposomal amphotericin B is the drug of choice. Nevertheless, limited safety data for the use of liposomal amphotericin B in neonates are available. Safety and efficacy of voriconazole were evaluated on 31 paediatric patients treated for 6–9 weeks for invasive aspergillosis in a prospective study showing that voriconazole is generally effective in paediatric patients, with a favourable risk–benefit balance and an overall safety profile similar to adults [31]. However, there is no firm data on the dosing of voriconazole in children aged 2 to 12 years.
In a large multicenter randomized trial on 277 patients older than 12 years old comparing children treated with voriconazole to the ones treated with amphotericin B, the first group was found to have a higher response rate, better survival at 12 weeks of treatment and fewer adverse events [22]. Nevertheless, there is no clear evidence to prefer one of the two drugs, since there is a lack of controlled and randomized comparison studies. On the other hand, there is an unequivocal need to monitor voriconazole plasma levels during therapy, especially in younger patients requiring higher doses [25]; plasma concentrations of 1—5 mg/l are usually considered adequate for prophylaxis and treatment of IPA. The dosages of most common antifungal drugs used for paediatric invasive aspergillosis are summarized in Table 4. Lastly, a few studies have investigated the safety and efficacy of drug combination in children; in a small retrospective study in leukemic patients with invasive fungal disease, 9 of which with invasive aspergillosis, the combination of voriconazole and caspofungin was safe and effective but further studies are needed [27]. Data providing evidence for biologic drug use in ABPA are scarce at the moment [37].
Table 4
Dosages of most common antifungal drugs in paediatric IPA
Drug
Dosage by age group
Adverse events
Newborns
1–24 months
2–12 years
13–18 years
Voriconazole e.v (mg/Kg/die)
Not approved
Not approved
14 (in 2 doses)
8 (in 2 doses)
Visual disturbances
Hepatotoxicity
Hypersensitivity
Skin rash
Voriconazole per os (mg/die)
Not approved
Not approved
400 (in 2 doses)
400 (in 2 doses)
Conventional amphotericin e.v (mg/Kg/die)
1–1,5
1–1,5
1–1,5
1–1,5
Nephrotoxicity
Electrolyte disturbances
Hypersensitivity
Amphotericin lipid formulation (mg/Kg/die)
5
3–5
NA
5
3–5
3–4
5
3–5
3–4
5
3–5
3–4
Mild nephrotoxicity
Electrolyte disturbances
Caspofungin (mg/m2/die)
25
50 (max 70); 70 per day
1
50 (max 70); 70 per day
1
50 (max 70); 70 per day
1
Fever
Hepatotoxicity
Cutaneous rash Tachycardia
Headache
Posaconazole per os (mg/die)
NA
NA
NA
800 (in 2 o 4 doses)
Hepatotoxicity
Nausea and vomiting
Headache
Itraconazole per os (mg/Kg/die)
NA
NA
5 (in 2 doses)
5 (in 2 doses)
Abdominal pain
Nausea
Skin rash
Abbreviations: NA Not available, BLC Amphotericin B lipid complex, L-AMB Amphotericin B liposomal, ABCD Amphotericin B colloidal dispersion

What is the optimal duration of treatment for IPA in children?

The duration of antifungal therapy has not been established and often needs to be individualized according to the child immune status. In a paediatric population examined in 2007, the mean duration of therapy was 93 days (range 1–880) with a partial response in 45% of cases. However, cases of severely immune-compromised patients in whom therapy was continued indefinitely have also been described [38]. In the prospective study of Martin et al., patients were treated for 6–9 weeks according to clinical response [31]. The most relevant issues in paediatric patients are the sequence of drugs to be used and the clinical parameters to be followed to evaluate the efficacy of therapy. Two main RCTs have been performed in the past in paediatric age groups [29, 39]. Although the expected outcomes are different, they agree that clinical improvement and fever relief for at least three consecutive days are the best evidence of therapy success. Even if regular dosing of GM antigen, after a first positive determination, could be helpful to guide antifungal therapy duration, a negative finding is not sufficient to discontinue therapy [13].

What are the main prophylactic drugs used for IPA in children?

Most of the evidence available in adults suggests the use of posaconazole for prophylactic purpose. It can be used in patients aged older than 13 years, neutropenic, with de novo or recurrent forms of AML, recurrent forms of acute lymphoblastic leukemia, stem cell transplant recipients or affected by GVHD [16] oral voriconazole or itraconazole are the drugs of choice for children aged 2 to 12 years [16, 23, 30]. In patients younger than 2 years of age or unable to take oral drugs, liposomal amphotericin B may be used [15], or, in those older than 2 years, intravenous voriconazole may be prescribed [39]. In a multicenter RCT including 517 patients from 3 months to 30 years of age, caspofungin was found to be superior to fluconazole in reducing invasive fungal diseases, including invasive aspergillosis [18].

Conclusions

Invasive pulmonary aspergillosis is a life-threatening condition and one of the leading causes of morbidity and mortality in fragile patients, however its diagnosis and management continue to be a clinical challenge. Among children, haematological malignancies, a previous organ transplant, and other primary or acquired immunodeficiency are the main risk factors for IPA.
GM antigen detection is a first-line diagnostic tool for high-risk patients with suspected aspergillosis and consecutive tests on BAL provide a high PPV, especially in a compatible clinical and radiological picture. No solid data regarding β-D-glucan diagnostic role in children are available, and PCR assays are not standardized nor validated for Aspergillus spp., therefore those tests should not be routinely used.
Voriconazole is currently the drug of choice in children older than 2 years. Liposomal amphotericin B should be administered in children younger than 2 years old or if voriconazole is contraindicated,. An unequivocal recommendation on the duration of treatment is missing since literature data are inconsistent. Clinical status and inflammatory and microbiological findings can guide an individualized therapy length. Voriconazole or itraconazole is suggested in children younger than 13 years old needing antifungal prophylaxis due to the paucity of safety data regarding posaconazole, while it can be safely used in older children.
Data about the diagnosis and management of IPA in children and adolescents are scarce, and no significant progress has been made in the last decades, suggesting that further high-quality studies are needed to improve clinical strategies.

Acknowledgements

Not applicable.

Declarations

Not applicable.
Not applicable.

Competing interests

The authors declare that they have no competing of interests.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat Kimberlin D, Barnett ED, Lynfield R, American SMH, Academy of Pediatrics. Aspergillosis. Red Book. Red Book,. Report of the Commitee on Infectious Disease. Itasca. 2021;2021:211–5. Kimberlin D, Barnett ED, Lynfield R, American SMH, Academy of Pediatrics. Aspergillosis. Red Book. Red Book,. Report of the Commitee on Infectious Disease. Itasca. 2021;2021:211–5.
2.
Zurück zum Zitat Manti S, Parisi GF, Papale M, Licari A, Chiappini E, Mulè E, et al. Allergic bronchopulmonary aspergillosis in children. Pediatr Allergy Immunol. 2020;31:20–2.CrossRefPubMed Manti S, Parisi GF, Papale M, Licari A, Chiappini E, Mulè E, et al. Allergic bronchopulmonary aspergillosis in children. Pediatr Allergy Immunol. 2020;31:20–2.CrossRefPubMed
3.
Zurück zum Zitat Page MJ, McKenzie JE, Bossuyt PM, Boutron I, Hoffmann TC, Mulrow CD, et al. The PRISMA 2020 statement: an updated guideline for reporting systematic reviews. BMJ. 2021;372:n71. Page MJ, McKenzie JE, Bossuyt PM, Boutron I, Hoffmann TC, Mulrow CD, et al. The PRISMA 2020 statement: an updated guideline for reporting systematic reviews. BMJ. 2021;372:n71.
4.
Zurück zum Zitat Jadad AR, Moore RA, Carroll D, Jenkinson C, Reynolds DJ, Gavaghan DJ, et al. Assessing the quality of reports of randomized clinical trials: is blinding necessary? Control Clin Trials. 1996;17:1–12.CrossRefPubMed Jadad AR, Moore RA, Carroll D, Jenkinson C, Reynolds DJ, Gavaghan DJ, et al. Assessing the quality of reports of randomized clinical trials: is blinding necessary? Control Clin Trials. 1996;17:1–12.CrossRefPubMed
5.
Zurück zum Zitat Slim K, Nini E, Forestier D, Kwiatkowski F, Panis Y, Chipponi J. Methodological index for non-randomized studies (minors): development and validation of a new instrument. ANZ J Surg. 2003;73:712–6.CrossRefPubMed Slim K, Nini E, Forestier D, Kwiatkowski F, Panis Y, Chipponi J. Methodological index for non-randomized studies (minors): development and validation of a new instrument. ANZ J Surg. 2003;73:712–6.CrossRefPubMed
6.
Zurück zum Zitat von Elm E, Altman DG, Egger M, Pocock SJ, Gøtzsche PC, Vandenbroucke JP. The Strengthening the Reporting of Observational Studies in Epidemiology (STROBE) statement: guidelines for reporting observational studies. Lancet Elsevier. 2007;370:1453–7.CrossRef von Elm E, Altman DG, Egger M, Pocock SJ, Gøtzsche PC, Vandenbroucke JP. The Strengthening the Reporting of Observational Studies in Epidemiology (STROBE) statement: guidelines for reporting observational studies. Lancet Elsevier. 2007;370:1453–7.CrossRef
7.
Zurück zum Zitat Anantasit N, Nuntacharruksa N, Incharoen P, Preutthipan A. Clinical and Pathological Correlation in Pediatric Invasive Pulmonary Aspergillosis. Front Pediatr. 2018;6:31.CrossRefPubMedPubMedCentral Anantasit N, Nuntacharruksa N, Incharoen P, Preutthipan A. Clinical and Pathological Correlation in Pediatric Invasive Pulmonary Aspergillosis. Front Pediatr. 2018;6:31.CrossRefPubMedPubMedCentral
8.
Zurück zum Zitat Avcu G, Karapinar DY, Akinci AB, Sivis ZO, Sahin A, Bal ZS, et al. Utility of the serum galactomannan assay for the diagnosis of invasive aspergillosis in children with acute lymphoblastic leukemia. Int J Infect Dis IJID Off Publ Int Soc Infect Dis. 2017;54:8–12. Avcu G, Karapinar DY, Akinci AB, Sivis ZO, Sahin A, Bal ZS, et al. Utility of the serum galactomannan assay for the diagnosis of invasive aspergillosis in children with acute lymphoblastic leukemia. Int J Infect Dis IJID Off Publ Int Soc Infect Dis. 2017;54:8–12.
9.
Zurück zum Zitat Badiee P, Alborzi A, Karimi M, Pourabbas B, Haddadi P, Mardaneh J, et al. Diagnostic potential of nested PCR, galactomannan EIA, and beta-D-glucan for invasive aspergillosis in pediatric patients. J Infect Dev Ctries. 2012;6:352–7.CrossRefPubMed Badiee P, Alborzi A, Karimi M, Pourabbas B, Haddadi P, Mardaneh J, et al. Diagnostic potential of nested PCR, galactomannan EIA, and beta-D-glucan for invasive aspergillosis in pediatric patients. J Infect Dev Ctries. 2012;6:352–7.CrossRefPubMed
10.
Zurück zum Zitat Burgos A, Zaoutis TE, Dvorak CC, Hoffman JA, Knapp KM, Nania JJ, et al. Pediatric invasive aspergillosis: a multicenter retrospective analysis of 139 contemporary cases. Pediatrics. 2008;121:e1286-1294.CrossRefPubMed Burgos A, Zaoutis TE, Dvorak CC, Hoffman JA, Knapp KM, Nania JJ, et al. Pediatric invasive aspergillosis: a multicenter retrospective analysis of 139 contemporary cases. Pediatrics. 2008;121:e1286-1294.CrossRefPubMed
11.
Zurück zum Zitat Çağlar İ, Özkerim D, Tahta N, Düzgöl M, Bayram N, Demirağ B, et al. Assessment of Serum Galactomannan Test Results of Pediatric Patients With Hematologic Malignancies According to Consecutive Positivity and Threshold Level in Terms of Invasive Aspergillosis Diagnosis: Cross-Sectional Research in a Tertiary Care Hospital. J Pediatr Hematol Oncol. 2020;42:e271–6.CrossRefPubMed Çağlar İ, Özkerim D, Tahta N, Düzgöl M, Bayram N, Demirağ B, et al. Assessment of Serum Galactomannan Test Results of Pediatric Patients With Hematologic Malignancies According to Consecutive Positivity and Threshold Level in Terms of Invasive Aspergillosis Diagnosis: Cross-Sectional Research in a Tertiary Care Hospital. J Pediatr Hematol Oncol. 2020;42:e271–6.CrossRefPubMed
12.
Zurück zum Zitat Choi S-H, Kang E-S, Eo H, Yoo S-Y, Kim JH, Yoo KH, et al. Aspergillus galactomannan antigen assay and invasive aspergillosis in pediatric cancer patients and hematopoietic stem cell transplant recipients. Pediatr Blood Cancer. 2013;60:316–22.CrossRefPubMed Choi S-H, Kang E-S, Eo H, Yoo S-Y, Kim JH, Yoo KH, et al. Aspergillus galactomannan antigen assay and invasive aspergillosis in pediatric cancer patients and hematopoietic stem cell transplant recipients. Pediatr Blood Cancer. 2013;60:316–22.CrossRefPubMed
13.
Zurück zum Zitat de Mol M, de Jongste JC, van Westreenen M, Merkus PJFM, de Vries AHC, Hop WCJ, et al. Diagnosis of invasive pulmonary aspergillosis in children with bronchoalveolar lavage galactomannan. Pediatr Pulmonol. 2013;48:789–96.CrossRefPubMed de Mol M, de Jongste JC, van Westreenen M, Merkus PJFM, de Vries AHC, Hop WCJ, et al. Diagnosis of invasive pulmonary aspergillosis in children with bronchoalveolar lavage galactomannan. Pediatr Pulmonol. 2013;48:789–96.CrossRefPubMed
14.
Zurück zum Zitat Dinand V, Anjan M, Oberoi JK, Khanna S, Yadav SP, Wattal C, et al. Threshold of galactomannan antigenemia positivity for early diagnosis of invasive aspergillosis in neutropenic children. J Microbiol Immunol Infect Wei Mian Yu Gan Ran Za Zhi. 2016;49:66–73.CrossRefPubMed Dinand V, Anjan M, Oberoi JK, Khanna S, Yadav SP, Wattal C, et al. Threshold of galactomannan antigenemia positivity for early diagnosis of invasive aspergillosis in neutropenic children. J Microbiol Immunol Infect Wei Mian Yu Gan Ran Za Zhi. 2016;49:66–73.CrossRefPubMed
15.
Zurück zum Zitat Döring M, Hartmann U, Erbacher A, Lang P, Handgretinger R, Müller I. Caspofungin as antifungal prophylaxis in pediatric patients undergoing allogeneic hematopoietic stem cell transplantation: a retrospective analysis. BMC Infect Dis. 2012;12:151.CrossRefPubMedPubMedCentral Döring M, Hartmann U, Erbacher A, Lang P, Handgretinger R, Müller I. Caspofungin as antifungal prophylaxis in pediatric patients undergoing allogeneic hematopoietic stem cell transplantation: a retrospective analysis. BMC Infect Dis. 2012;12:151.CrossRefPubMedPubMedCentral
16.
Zurück zum Zitat Döring M, Eikemeier M, Cabanillas Stanchi KM, Hartmann U, Ebinger M, Schwarze C-P, et al. Antifungal prophylaxis with posaconazole vs fluconazole or itraconazole in pediatric patients with neutropenia. Eur J Clin Microbiol Infect Dis Off Publ Eur Soc Clin Microbiol. 2015;34:1189–200.CrossRef Döring M, Eikemeier M, Cabanillas Stanchi KM, Hartmann U, Ebinger M, Schwarze C-P, et al. Antifungal prophylaxis with posaconazole vs fluconazole or itraconazole in pediatric patients with neutropenia. Eur J Clin Microbiol Infect Dis Off Publ Eur Soc Clin Microbiol. 2015;34:1189–200.CrossRef
17.
Zurück zum Zitat Fisher BT, Zaoutis TE, Park JR, Bleakley M, Englund JA, Kane C, et al. Galactomannan Antigen Testing for Diagnosis of Invasive Aspergillosis in Pediatric Hematology Patients. J Pediatr Infect Dis Soc. 2012;1:103–11.CrossRef Fisher BT, Zaoutis TE, Park JR, Bleakley M, Englund JA, Kane C, et al. Galactomannan Antigen Testing for Diagnosis of Invasive Aspergillosis in Pediatric Hematology Patients. J Pediatr Infect Dis Soc. 2012;1:103–11.CrossRef
18.
Zurück zum Zitat Fisher BT, Zaoutis T, Dvorak CC, Nieder M, Zerr D, Wingard JR, et al. Effect of Caspofungin vs Fluconazole Prophylaxis on Invasive Fungal Disease Among Children and Young Adults With Acute Myeloid Leukemia: A Randomized Clinical Trial. JAMA. 2019;322:1673–81.CrossRefPubMedPubMedCentral Fisher BT, Zaoutis T, Dvorak CC, Nieder M, Zerr D, Wingard JR, et al. Effect of Caspofungin vs Fluconazole Prophylaxis on Invasive Fungal Disease Among Children and Young Adults With Acute Myeloid Leukemia: A Randomized Clinical Trial. JAMA. 2019;322:1673–81.CrossRefPubMedPubMedCentral
19.
Zurück zum Zitat Fisher BT, Westling T, Boge CLK, Zaoutis TE, Dvorak CC, Nieder M, et al. Prospective Evaluation of Galactomannan and (1→3) β-d-Glucan Assays as Diagnostic Tools for Invasive Fungal Disease in Children, Adolescents, and Young Adults With Acute Myeloid Leukemia Receiving Fungal Prophylaxis. J Pediatr Infect Dis Soc. 2021;10:864–71.CrossRef Fisher BT, Westling T, Boge CLK, Zaoutis TE, Dvorak CC, Nieder M, et al. Prospective Evaluation of Galactomannan and (1→3) β-d-Glucan Assays as Diagnostic Tools for Invasive Fungal Disease in Children, Adolescents, and Young Adults With Acute Myeloid Leukemia Receiving Fungal Prophylaxis. J Pediatr Infect Dis Soc. 2021;10:864–71.CrossRef
20.
Zurück zum Zitat Gefen A, Zaidman I, Shachor-Meyouhas Y, Avidor I, Hakim F, Weyl Ben-Arush M, et al. Serum galactomannan screening for diagnosis of invasive pulmonary aspergillosis in children after stem cell transplantation or with high-risk leukemia. Pediatr Hematol Oncol. 2015;32:146–52.CrossRefPubMed Gefen A, Zaidman I, Shachor-Meyouhas Y, Avidor I, Hakim F, Weyl Ben-Arush M, et al. Serum galactomannan screening for diagnosis of invasive pulmonary aspergillosis in children after stem cell transplantation or with high-risk leukemia. Pediatr Hematol Oncol. 2015;32:146–52.CrossRefPubMed
21.
Zurück zum Zitat Han SB, Kim SK, Bae EY, Lee JW, Yoon J-S, Chung N-G, et al. Clinical Features and Prognosis of Invasive Pulmonary Aspergillosis in Korean Children with Hematologic/Oncologic Diseases. J Korean Med Sci. 2015;30:1121–8.CrossRefPubMedPubMedCentral Han SB, Kim SK, Bae EY, Lee JW, Yoon J-S, Chung N-G, et al. Clinical Features and Prognosis of Invasive Pulmonary Aspergillosis in Korean Children with Hematologic/Oncologic Diseases. J Korean Med Sci. 2015;30:1121–8.CrossRefPubMedPubMedCentral
22.
Zurück zum Zitat Herbrecht R, Denning DW, Patterson TF, Bennett JE, Greene RE, Oestmann J-W, et al. Voriconazole versus amphotericin B for primary therapy of invasive aspergillosis. N Engl J Med. 2002;347:408–15.CrossRefPubMed Herbrecht R, Denning DW, Patterson TF, Bennett JE, Greene RE, Oestmann J-W, et al. Voriconazole versus amphotericin B for primary therapy of invasive aspergillosis. N Engl J Med. 2002;347:408–15.CrossRefPubMed
23.
Zurück zum Zitat Itsaradisaikul S, Pakakasama S, Boonsathorn S, Techasaensiri C, Rattanasiri S, Apiwattanakul N. Invasive Fungal Disease Among Pediatric and Adolescent Patients Undergoing Itraconazole Prophylaxis After Hematopoietic Stem Cell Transplantation. Transplant Proc. 2021;53:2021–8.CrossRefPubMed Itsaradisaikul S, Pakakasama S, Boonsathorn S, Techasaensiri C, Rattanasiri S, Apiwattanakul N. Invasive Fungal Disease Among Pediatric and Adolescent Patients Undergoing Itraconazole Prophylaxis After Hematopoietic Stem Cell Transplantation. Transplant Proc. 2021;53:2021–8.CrossRefPubMed
24.
Zurück zum Zitat Jha AK, Bansal D, Chakrabarti A, Shivaprakash MR, Trehan A, Marwaha RK. Serum galactomannan assay for the diagnosis of invasive aspergillosis in children with haematological malignancies. Mycoses. 2013;56:442–8.CrossRefPubMed Jha AK, Bansal D, Chakrabarti A, Shivaprakash MR, Trehan A, Marwaha RK. Serum galactomannan assay for the diagnosis of invasive aspergillosis in children with haematological malignancies. Mycoses. 2013;56:442–8.CrossRefPubMed
25.
Zurück zum Zitat Kato K, Nagao M, Yamamoto M, Matsumura Y, Takakura S, Fukuda K, et al. Oral administration and younger age decrease plasma concentrations of voriconazole in pediatric patients. J Infect Chemother Off J Jpn Soc Chemother. 2016;22:27–31.CrossRef Kato K, Nagao M, Yamamoto M, Matsumura Y, Takakura S, Fukuda K, et al. Oral administration and younger age decrease plasma concentrations of voriconazole in pediatric patients. J Infect Chemother Off J Jpn Soc Chemother. 2016;22:27–31.CrossRef
26.
Zurück zum Zitat Kazakou N, Vyzantiadis TA, Gambeta A, Vasileiou E, Tsotridou E, Kotsos D, et al. Invasive fungal infections in a pediatric hematology-oncology department: A 16-year retrospective study. Curr Med Mycol. 2020;6:37–42.PubMedPubMedCentral Kazakou N, Vyzantiadis TA, Gambeta A, Vasileiou E, Tsotridou E, Kotsos D, et al. Invasive fungal infections in a pediatric hematology-oncology department: A 16-year retrospective study. Curr Med Mycol. 2020;6:37–42.PubMedPubMedCentral
27.
Zurück zum Zitat Lee KH, Lim YT, Hah JO, Kim YK, Lee CH, Lee JM. Voriconazole plus caspofungin for treatment of invasive fungal infection in children with acute leukemia. Blood Res. 2017;52:167–73.CrossRefPubMedPubMedCentral Lee KH, Lim YT, Hah JO, Kim YK, Lee CH, Lee JM. Voriconazole plus caspofungin for treatment of invasive fungal infection in children with acute leukemia. Blood Res. 2017;52:167–73.CrossRefPubMedPubMedCentral
28.
Zurück zum Zitat Loeffler J, Hafner J, Mengoli C, Wirth C, Heussel CP, Löffler C, et al. Prospective Biomarker Screening for Diagnosis of Invasive Aspergillosis in High-Risk Pediatric Patients. J Clin Microbiol. 2017;55:101–9.CrossRefPubMed Loeffler J, Hafner J, Mengoli C, Wirth C, Heussel CP, Löffler C, et al. Prospective Biomarker Screening for Diagnosis of Invasive Aspergillosis in High-Risk Pediatric Patients. J Clin Microbiol. 2017;55:101–9.CrossRefPubMed
29.
Zurück zum Zitat Maertens JA, Madero L, Reilly AF, Lehrnbecher T, Groll AH, Jafri HS, et al. A randomized, double-blind, multicenter study of caspofungin versus liposomal amphotericin B for empiric antifungal therapy in pediatric patients with persistent fever and neutropenia. Pediatr Infect Dis J. 2010;29:415–20.CrossRefPubMed Maertens JA, Madero L, Reilly AF, Lehrnbecher T, Groll AH, Jafri HS, et al. A randomized, double-blind, multicenter study of caspofungin versus liposomal amphotericin B for empiric antifungal therapy in pediatric patients with persistent fever and neutropenia. Pediatr Infect Dis J. 2010;29:415–20.CrossRefPubMed
30.
Zurück zum Zitat Maron GM, Hayden RT, Rodriguez A, Rubnitz JE, Flynn PM, Shenep JL, et al. Voriconazole prophylaxis in children with cancer: changing outcomes and epidemiology of fungal infections. Pediatr Infect Dis J. 2013;32:e451-455.CrossRefPubMed Maron GM, Hayden RT, Rodriguez A, Rubnitz JE, Flynn PM, Shenep JL, et al. Voriconazole prophylaxis in children with cancer: changing outcomes and epidemiology of fungal infections. Pediatr Infect Dis J. 2013;32:e451-455.CrossRefPubMed
31.
Zurück zum Zitat Martin JM, Macias-Parra M, Mudry P, Conte U, Yan JL, Liu P, et al. Safety, Efficacy, and Exposure-Response of Voriconazole in Pediatric Patients With Invasive Aspergillosis, Invasive Candidiasis or Esophageal Candidiasis. Pediatr Infect Dis J. 2017;36:e1-13.CrossRefPubMed Martin JM, Macias-Parra M, Mudry P, Conte U, Yan JL, Liu P, et al. Safety, Efficacy, and Exposure-Response of Voriconazole in Pediatric Patients With Invasive Aspergillosis, Invasive Candidiasis or Esophageal Candidiasis. Pediatr Infect Dis J. 2017;36:e1-13.CrossRefPubMed
32.
Zurück zum Zitat Mohammadi S, Khalilzadeh S, Goudarzipour K, Hassanzad M, Mahdaviani A, Aarabi N, et al. Bronchoalveolar galactomannan in invasive pulmonary aspergillosis: a prospective study in pediatric patients. Med Mycol. 2015;53:709–16.CrossRefPubMed Mohammadi S, Khalilzadeh S, Goudarzipour K, Hassanzad M, Mahdaviani A, Aarabi N, et al. Bronchoalveolar galactomannan in invasive pulmonary aspergillosis: a prospective study in pediatric patients. Med Mycol. 2015;53:709–16.CrossRefPubMed
33.
Zurück zum Zitat Qiu K-Y, Liao X-Y, Huang K, Xu H-G, Li Y, Fang J-P, et al. The early diagnostic value of serum galactomannan antigen test combined with chest computed tomography for invasive pulmonary aspergillosis in pediatric patients after hematopoietic stem cell transplantation. Clin Transplant. 2019;33:e13641.CrossRefPubMed Qiu K-Y, Liao X-Y, Huang K, Xu H-G, Li Y, Fang J-P, et al. The early diagnostic value of serum galactomannan antigen test combined with chest computed tomography for invasive pulmonary aspergillosis in pediatric patients after hematopoietic stem cell transplantation. Clin Transplant. 2019;33:e13641.CrossRefPubMed
34.
Zurück zum Zitat Vrioni G, Theodoridou K, Tsiamis C, Mavrouli M, Kapsimali V, Rigopoulos D, et al. Use of Galactomannan Antigen and Aspergillus DNA Real-time Polymerase Chain Reaction as Routine Methods for Invasive Aspergillosis in Immunosuppressed Children in Greece. Clin Ther. 2018;40:918-924.e2.CrossRefPubMed Vrioni G, Theodoridou K, Tsiamis C, Mavrouli M, Kapsimali V, Rigopoulos D, et al. Use of Galactomannan Antigen and Aspergillus DNA Real-time Polymerase Chain Reaction as Routine Methods for Invasive Aspergillosis in Immunosuppressed Children in Greece. Clin Ther. 2018;40:918-924.e2.CrossRefPubMed
35.
Zurück zum Zitat Zaoutis TE, Heydon K, Chu JH, Walsh TJ, Steinbach WJ. Epidemiology, outcomes, and costs of invasive aspergillosis in immunocompromised children in the United States, 2000. Pediatrics. 2006;117:e711-716.CrossRefPubMed Zaoutis TE, Heydon K, Chu JH, Walsh TJ, Steinbach WJ. Epidemiology, outcomes, and costs of invasive aspergillosis in immunocompromised children in the United States, 2000. Pediatrics. 2006;117:e711-716.CrossRefPubMed
36.
Zurück zum Zitat Donnelly JP, Chen SC, Kauffman CA, Steinbach WJ, Baddley JW, Verweij PE, et al. Revision and Update of the Consensus Definitions of Invasive Fungal Disease From the European Organization for Research and Treatment of Cancer and the Mycoses Study Group Education and Research Consortium. Clin Infect Dis. 2020;71:1367–76.CrossRefPubMed Donnelly JP, Chen SC, Kauffman CA, Steinbach WJ, Baddley JW, Verweij PE, et al. Revision and Update of the Consensus Definitions of Invasive Fungal Disease From the European Organization for Research and Treatment of Cancer and the Mycoses Study Group Education and Research Consortium. Clin Infect Dis. 2020;71:1367–76.CrossRefPubMed
37.
Zurück zum Zitat Manti S, Giallongo A, Parisi GF, Papale M, Mulè E, Aloisio D, et al. Biologic drugs in treating allergic bronchopulmonary aspergillosis in patients with cystic fibrosis: a systematic review. Eur Respir Rev Off J Eur Respir Soc. 2022;31:220011.CrossRef Manti S, Giallongo A, Parisi GF, Papale M, Mulè E, Aloisio D, et al. Biologic drugs in treating allergic bronchopulmonary aspergillosis in patients with cystic fibrosis: a systematic review. Eur Respir Rev Off J Eur Respir Soc. 2022;31:220011.CrossRef
38.
Zurück zum Zitat Krishna G, Sansone-Parsons A, Martinho M, Kantesaria B, Pedicone L. Posaconazole plasma concentrations in juvenile patients with invasive fungal infection. Antimicrob Agents Chemother. 2007;51:812–8.CrossRefPubMedPubMedCentral Krishna G, Sansone-Parsons A, Martinho M, Kantesaria B, Pedicone L. Posaconazole plasma concentrations in juvenile patients with invasive fungal infection. Antimicrob Agents Chemother. 2007;51:812–8.CrossRefPubMedPubMedCentral
39.
Zurück zum Zitat Prentice HG, Hann IM, Herbrecht R, Aoun M, Kvaloy S, Catovsky D, et al. A randomized comparison of liposomal versus conventional amphotericin B for the treatment of pyrexia of unknown origin in neutropenic patients. Br J Haematol. 1997;98:711–8.CrossRefPubMed Prentice HG, Hann IM, Herbrecht R, Aoun M, Kvaloy S, Catovsky D, et al. A randomized comparison of liposomal versus conventional amphotericin B for the treatment of pyrexia of unknown origin in neutropenic patients. Br J Haematol. 1997;98:711–8.CrossRefPubMed
Metadaten
Titel
Management of pulmonary aspergillosis in children: a systematic review
verfasst von
Vito Terlizzi
Marco Antonio Motisi
Roberta Pellegrino
Luisa Galli
Giovanni Taccetti
Elena Chiappini
Publikationsdatum
01.12.2023
Verlag
BioMed Central
Erschienen in
Italian Journal of Pediatrics / Ausgabe 1/2023
Elektronische ISSN: 1824-7288
DOI
https://doi.org/10.1186/s13052-023-01440-9

Weitere Artikel der Ausgabe 1/2023

Italian Journal of Pediatrics 1/2023 Zur Ausgabe

Ähnliche Überlebensraten nach Reanimation während des Transports bzw. vor Ort

29.05.2024 Reanimation im Kindesalter Nachrichten

Laut einer Studie aus den USA und Kanada scheint es bei der Reanimation von Kindern außerhalb einer Klinik keinen Unterschied für das Überleben zu machen, ob die Wiederbelebungsmaßnahmen während des Transports in die Klinik stattfinden oder vor Ort ausgeführt werden. Jedoch gibt es dabei einige Einschränkungen und eine wichtige Ausnahme.

Alter der Mutter beeinflusst Risiko für kongenitale Anomalie

28.05.2024 Kinder- und Jugendgynäkologie Nachrichten

Welchen Einfluss das Alter ihrer Mutter auf das Risiko hat, dass Kinder mit nicht chromosomal bedingter Malformation zur Welt kommen, hat eine ungarische Studie untersucht. Sie zeigt: Nicht nur fortgeschrittenes Alter ist riskant.

Begünstigt Bettruhe der Mutter doch das fetale Wachstum?

Ob ungeborene Kinder, die kleiner als die meisten Gleichaltrigen sind, schneller wachsen, wenn die Mutter sich mehr ausruht, wird diskutiert. Die Ergebnisse einer US-Studie sprechen dafür.

Bei Amblyopie früher abkleben als bisher empfohlen?

22.05.2024 Fehlsichtigkeit Nachrichten

Bei Amblyopie ist das frühzeitige Abkleben des kontralateralen Auges in den meisten Fällen wohl effektiver als der Therapiestandard mit zunächst mehrmonatigem Brilletragen.

Update Pädiatrie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.