Skip to main content
Erschienen in: Infectious Diseases and Therapy 1/2018

Open Access 22.12.2017 | Review

Challenges and Solution of Invasive Aspergillosis in Non-neutropenic Patients: A Review

verfasst von: Matteo Bassetti, Maddalena Peghin, Antonio Vena

Erschienen in: Infectious Diseases and Therapy | Ausgabe 1/2018

Abstract

Invasive aspergillosis (IA) is a serious opportunistic infection, which has increasingly been recognized as an emerging disease of non-neutropenic patients. In this group of patients, the diagnosis of IA can be challenging owing to the lack of specificity of symptoms, the difficulty in discriminating colonization from infection, and the lower sensitivity of microbiological and radiological tests compared with immunocompromised patients. The aim of this article is to present to clinicians a critical review on the management of IA in non-neutropenic patients.
Hinweise

Enhanced Content

To view enhanced content for this article go to http://​www.​medengine.​com/​Redeem/​E10DF0602F023114​.

Introduction

Invasive aspergillosis (IA) is a serious opportunistic infection that continues to increase its incidence in immunosuppressed or hospitalized patients with severe underlying diseases, with high rates of morbidity and mortality [1]. Over the past decades, the population of patients susceptible to develop IA has expanded significantly and IA has increasingly been recognized as an emerging disease of non-neutropenic patients with an incidence varying between 0.33–5.8% [2, 3]. In addition, the classical view of IA has been modified, since various clinical syndromes have been considered as a continuous spectrum of the disease, whose manifestations are defined by the complex interaction between pathogen and host factors [4].
Invasive aspergillosis in non-neutropenic patients is associated with bad prognosis, with mortality rates exceeding 80%, mainly due to delayed diagnosis [2, 3]. Difficulties in achieving a timely diagnosis of IA in non-neutropenic patients is related to the non-specificity of clinical presentation and to lower yields with diagnostic tests compared to neutropenic patients [2, 3].
The aim of this article is to present to clinicians a critical review on the recognition, risk factors, microbiological and radiological diagnosis, and management of IA in non-neutropenic patients.

Compliance with Ethical Guidelines

This article is based on previously conducted studies and does not involve any studies of human or animal subjects performed by any of the authors.

Host Risk Factors and Spectrum of Disease

Invasive aspergillosis has been traditionally considered as an opportunistic infection mainly occurring in patients with well-established risk factors, such as neutropenia, hematologic malignancies, allogeneic bone marrow transplantation, solid organ transplantation, solid cancer or HIV [5].
However, an increasing number of reports have shown that Aspergillus spp. can cause invasive disease in other categories of non-neutropenic patients, including those with severe chronic obstructive pulmonary disease (COPD) requiring high-dose steroid therapy, with Child–Pugh C hepatic cirrhosis, and systemic diseases requiring immunosuppressive therapy, including new monoclonal agents in patients with autoimmune diseases [6].
An emerging broad group of patients who are admitted to the intensive care unit (ICU) may also be susceptible to IA [3, 7], with previously reported rates varying widely from 0.017% to as high as 6.9% [6, 8]. In addition to host underlying conditions (Table 1) [6], immunoregulatory abnormalities following critical illness can induce a state of immunoparalysis, hampering adequate host response to fungal disease in the ICU [9]. Other predisposing risk factors frequently met in ICUs include acute respiratory distress syndrome (ARDS), severe sepsis, acute renal failure, and H1N1 virus infection (especially if CS prior to ICU admission) [8]. Moreover, environmental factors including climatic variables, airborne mold concentrations, geographic area, remodeling or construction works and environmental quality of the air may influence IA development [1].
Table 1
Risk of invasive aspergillosis among patients admitted to the intensive care unit by Meersseman et al. [6]
High-risk category
 Neutropenia (neutrophil count, !500 neutrophils/mm3)
 Hematological malignancy
 Allogeneic bone marrow transplantation
Intermediate-risk category
 Prolonged treatment with corticosteroids before admission to the ICU
 Autologous bone marrow transplantation
 Chronic obstructive pulmonary disease
 Liver cirrhosis with a duration of stay in the ICU > 7 days
 Solid-organ cancer HIV infection
 Lung transplantation
 Systemic diseases requiring immunosuppressive therapy
Low-risk category
 Severe burns
 Other solid-organ transplant recipients (e.g., heart, kidney, or liver transplant recipients)
 Steroid treatment with a duration of < 7 days
 Prolonged stay in the ICU (> 21 days)
 Malnutrition
 Post-cardiac surgery status
The nature of the immune suppression (the degree, duration, and type of immunodeficiency) influences the pathogenesis of disease. IA thus manifests as a spectrum of disease involving predominantly airway (tracheobronchitis), lung, or both. Another factor that makes IA in non-neutropenic patients difficult to diagnose is its non-specific symptomatology that makes clinical manifestations of IA (e.g., fever, cough, purulent sputum) indistinguishable from other bacterial bronchopneumonia [10]. Notably in previous studies, non-neutropenic patients have shown to be less symptomatic than neutropenic patients regarding fever as well as cough and chest pain [3].
In this setting, clinical diagnosis of IA is a challenge, and initiation of additional diagnostic examinations is often delayed because clinical suspicion is low. Standard diagnostic definitions by the European Organization for Research and Treatment of Cancer (EORTC) have been developed but have been validated only for patients with cancer or after hematopoietic stem cell transplants and cannot be extrapolated to non-neutropenic patients [11]. Blot and colleagues validated a clinical diagnostic algorithm that aims to discriminate colonization from probable IA in ICU patients with Aspergillus-positive endotracheal aspirate cultures [12].

Microbiological Diagnosis

The suspicion of invasive aspergillosis in non-neutropenic patients may be delayed because paucisymptomatic disease is not uncommon [3, 13, 14]. Another factor that makes invasive aspergillosis in non-neutropenic patients difficult to diagnose is its non-specific symptomatology that makes clinical manifestations of IA (e.g., fever, cough, purulent sputum) indistinguishable from other bacterial bronchopneumonia [15]. A high index of suspicion is required to successfully achieve a positive diagnosis, and it is advisable to perform fungal cultures and non-culture-based methods in all patients with relevant risk factors who present with an infectious complication. This recommendation would apply even if other agents had already been isolated or even during necropsy or because a positive culture result might have been achieved accidentally (Fig. 1).
The cultures of lower respiratory tract are easy and cheap and enable Aspergillus species to be identified and their antifungal susceptibility testing performed. However, cultures are slow and their yield in respiratory sample is notoriously low, with a sensitivity ranging between 20% and 50% [16, 17]. In addition, the clinical significance of isolating Aspergillus from respiratory samples remains unclear, because differentiating true infection from simple colonization can be difficult [18]. Once the fungus is detected in respiratory samples, experts in the field should interpret the isolation in the clinical context of the patient and eventually start antifungal therapy, if indicated.
In recent years, surrogate markers have been developed for diagnosis of IA, based on the detection of fungal cell wall component or fungal DNA in clinical specimens. The Platelia (Sanofi Diagnostic Pasterur, Marnes la Coquette, France) sandwich-enzyme immunosorbent assay (ELISA) for the detection of galactomannan (GM) is currently one of the more used methods. It is based on the detection of circulating antigens in biologic fluids, such as serum, urine, or BAL fluid [19, 20]. A meta-analysis including 27 studies reported an overall sensitivity of serum galactomannan assay of 71% and specificity of 89%. However, when onco-hematological patients were excluded from the analysis, the sensitivity and specificity of the test dropped to 22% and 84%, respectively [21].
In a recent prospective study of IA in non-neutropenic patients, Zhou et al. found a sensitivity of serum GM of 37.8% and a specificity of 87.1%, with a positive predictive value of only 60.8% [22]. Other specimens, such as bronchoalveolar lavage (BAL), have been proven to be more advantageous in the non-neutropenic population. At the index cutoff value of 0.5, the test yielded a sensitivity up to 100% and specificity ranging from 75% to 92% [22, 23]. Although in hematological patients the GM test may enable the early diagnosis of IA [24, 25] and monitoring the treatment response [2628], further studies are mandatory in non-neutropenic patients since these aspects remain to be determined in this population [29].
Another test is the 1-3-β-d-glucan assay, a polysaccharide component of the cell wall of many pathogenic fungi other than Mucoraceae and Cryptococcus. Four previous meta-analyses, mainly including patients with hematological disease, reported a good sensitivity but a very low specificity and positive predictive value for the diagnosis of fungal infection [3033]. In contrast, its negative predictive value was as high as 80–90%, thus making 1-3-β-d-glucan potentially useful to rule out the diagnosis of IA rather than to confirm it [3033].
However, because the significant heterogeneity in testing strategies, inclusion criteria, and low number of patients, the role of this biomarker in the diagnosis of IA in non-neutropenic patients is still unknown and future studies appear to be appropriate. In addition, detection is also limited by the high frequency of false-positive results including semi-syntetic β-lactam antibiotics, human blood components, cellulose hemodialysis, and exposure to gauze [34]. A few studies have evaluated the role of BDG in BAL, indicating a very low specificity and positive predictive value (≈ 20%) for IA in immunocompromised patients [35].
One test that reduces the time required to diagnose invasive aspergillosis is the amplification of genetic material from Aspergillus spp., which detects genetic sequences (18S rDNA, 28SrDNA, 5.8 SrDNA, mithocondrial DNA) in cultures as well as in direct clinical samples within a few hours [34]. Unfortunately, PCR is not yet universally standardized [36, 37], and cannot yet be included as a mycological criterion in the EORTC/MSG guidelines [11]. In addition, its usefulness in non-neutropenic patients is not yet clear, although the information that has been reported seems to be promising [3841]. Theoretically, the test should not be affected either by the immune status of the patients or by the presence of other fungal or bacterial pathogens.
With two positive consecutive results, this kind of test has a specificity close to 95% with a high positive likelihood ratio (LR 12.8) and should be considered highly indicative of an active Aspergillus spp. infection [37]; on the other hand, a single negative PCR result is sufficient to exclude the diagnosis [42]. In addition, when combined with other fungal biomarkers in serum (either GM or BDG) or in BAL (GM), Aspergillus PCR has shown to increase the diagnostic sensitivity up to 100%, further supporting the implementation of this technique in the revised definition of invasive fungal infection by the EORTC/MSG. Experience with non-neutropenic patients is scarce, but the efficacy of molecular techniques seems to be similar to that for the population with hematological malignancies [43]. Another application of molecular biology techniques for the diagnosis and treatment of invasive aspergillosis is the ability to detect azole-resistant strains earlier than do conventional methods [44]. Finally, it is important to mention the contribution of molecular techniques in genotyping fungal strains directly from clinical samples.
Recently, a lateral flow device (LFD), detecting a glycoprotein antigen found in the serum and BAL of patients with IA [45], has been proposed as a new point-of care diagnostic approach for also detecting IPA in non-hematological populations, including SOT and ICU patients [46, 47]. A recent multicenter study evaluating the LFD device in BAL from 133 ICU patients showed a sensitivity, specificity, and positive and negative predictive values for probable IA of 80%, 81%, 96%, and 44%, respectively [48]. However, despite such promising results, further and larger studies are warranted before safe conclusions on the performance of Aspergillus LDF can be reached. A multicenter study evaluating the role of Aspergillus LFD as an alternative to GM in BAL fluid is currently underway (clinicaltrial.gov identifier NCT 02058316).
Finally, different technologies detecting volatile organic compounds exhaled in the breath of patients infected with IA have recently been tested [49, 50], with a sensitivity ranging from 94% to 100% and specificity form 83% to 93% [49, 50]. Also, other tests including gliotoxin, bis(methylthio)gliotoxin, have been analyzed for diagnosing IA with interesting results [51, 52]. Despite this, their role in non-neutropenic patients remains to be clarified and additional analysis with a larger cohort of patients are needed.
In conclusion, diagnosis of invasive aspergillosis remains challenging because none of the available diagnostic tests provides sufficient sensitivity and specificity alone, so the optimal approach relies on the simultaneous performance of several diagnostic strategies, including cultures, fungal biomarkers and molecular tools.

Radiological Diagnosis

Pulmonary lesions caused by Aspergillus spp. can be responsible for a wide range of radiographic findings, as with disease manifestations potentially mutating depending upon the immune status of the host.
Chest radiographs are insensitive for detecting the earliest stages of pulmonary disease, but computed tomography (CT) scans typically demonstrate focal lesions. Several thoracic CT patterns are associated with pulmonary aspergillosis. Radiological patterns can be non-specific in non-neutropenic patients [9]. Radiographic signs of IA can vary from a single nodular lesions or larger masses to diffuse bilateral pulmonary infiltrates. The most typical imaging findings including the halo sign and the air crescent sign have shown high sensitivity (80%) and specificity (60–98%) in neutropenic patients. Nevertheless, both signs are uncommon, have a lower sensitivity (5–24%) and can be found even in non-infectious lesion processes in non-neutropenic patients [53].
A large study that included neutropenic and non-neutropenic patients showed that the more specific signs (nodules and cavitation) were infrequent, and that radiographic findings of consolidation, ground-glass infiltrates, and pleural effusions were seen more commonly [3]. In addition, many ICU patients have radiologic abnormalities masked by underlying acute processes (pleural effusion, atelectasis or ARDS) [9]. Of interest, airway-invasive or angio-invasive radiological patterns have been described in non-neutropenic heart transplant recipients with IA and have been associated with different presentations, time to diagnosis and mortality rates [13].
Given the current limitations of CT, efforts to improve diagnostic performance in pulmonary aspergillosis have been pursued. High-resolution CT pulmonary angiography (CTPA) can detect angio-invasion and vessel occlusion signs (VOS). VOS has been shown to be superior to classic CT signs observed in non-contrast enhanced studies to diagnose invasive pulmonary aspergillosis in immunocompromised patients, including those with hematologic malignancies and other causes, with a sensitivity of 0.94, specificity of 0.71 and a diagnostic odds-ratio of 36.8 [54].
Combined anatomic and functional imaging with 18F-fluorodeoxyglucose positron emission tomography (FDG-PET) is slowly gaining a foothold in the management of invasive fungal infections [55]. In current clinical practice, standalone FDG-PET/CT is not generally sufficient to differentiate IPA from malignancy or other active inflammatory lesions, such as active tuberculosis [55]. However, significant different FDG-PET/CT patterns in invasive and non-invasive forms of aspergillosis have been described [56]. Invasive aspergillosis usually presents with multiple hypermetabolic nodules and a higher peak (SUV 4.5; range 1.3–8.9), whereas non-invasive forms presents with isometabolic halo or nodule patterns with a relatively lower SUV peak value 1.6 (range 0.5–3.1) [56].
In real clinical practice, radiologic follow-up of aspergillosis is mostly carried out by serial CT scans and represents a challenge. FDG PET/CT has been found to be a valuable tool for early evaluation of treatment response in aspergillosis, particularly in patients with underlying pulmonary diseases (i.e., chronic obstructive pulmonary disease, previous TB, and cancer) and for establishing the appropriate treatment duration [57].
A recent study supported the use of a novel probe for detection of A. fumigatus lung infection based on antibody-guided positron emission tomography and magnetic resonance (immunoPET/MR). This promising imaging technique seems to allow accurate, non-invasive and rapid detection of fungal lung infection and discrimination of IPA from bacterial lung infections and general inflammatory responses [58].
In our opinion, the presence of a persistent pulmonary infection despite broad-spectrum antibiotics or abnormal thoracic imaging by CT scanning together with one of the host risk factors should trigger further diagnostic exploration through collection of respiratory secretions and/or laboratory markers. There is a potential role of FDG/PET-TC for radiological diagnosis and treatment monitoring in IA.

Treatment

Despite the many therapeutic options available today, the mortality rate of IA in non-neutropenic patients continues to be as high as 90% [6, 52], and is even higher than that reported in the hematological population, likely reflecting the lack of strategies of early diagnosis allowing early appropriate therapy in this population [1]. Indeed, in contrast to neutropenic patients, no consensus exists about the exact timeframe for starting empirical therapy in patients at high risk for Aspergillus infection and no microbiological evidence of IA [59].
In our opinion, non-neutropenic patients at high risk of IA (i.e., COPD, steroids and immunosuppressive therapy, hepatic failure, and ICU-related immunoparalysis) should receive adequate antifungal therapy upon suspicion of the fungal disease, even if a definitive proof of infection is still not obtained. Whenever possible, a CT scan, fungal cultures and a combination of serological biomarkers (GM, Aspergillus PCR and 1,3-b-D-glucan assay, as a screening strategy) should be performed and treatment should be revised and eventually withheld if the diagnosis of IA is not confirmed.
As for antifungal drugs, there has been considerable research in antifungal drugs targeted against IA over the past decade [16, 17, 6062]. To date, the antifungal agents licensed for the first-line treatment of IA include voriconazole and isavuconazole or amphotericin B and its lipid formulations [5]. The severity of the infection, the clinical form, renal or hepatic insufficiency, drug–drug interactions, requirement for therapeutic drug monitoring and its cost are some of the factors that can help in selecting the best drug.
During the last 10 years, voriconazole use has progressively become widespread. The largest randomized trial for primary therapy of invasive pulmonary aspergillosis demonstrated that voriconazole was superior to amphotericin B deoxycholate, followed by other licensed antifungal therapy [63]. At week 12, successful outcomes among non-neutropenic population (31 patients) were observed in 50.0% of the patients in the voriconazole group and in 31.6% of those in the amphotericin B group. Moreover, patients treated with voriconazole had significantly fewer adverse events that were drug-related, except for transient visual disturbances. Therefore, the authors of this study concluded that voriconazole was more beneficial for the treatment of IA than amphotericin B.
Other series studying non-neutropenic patients, with proven or probable invasive pulmonary aspergillosis, confirmed a favorable response rate with voriconazole [64, 65]. Particularly remarkable is one study of pulmonary and disseminated IA, including 103 non-neutropenic patients, in which receiving voriconazole treatment was found to be the only factor associated with a reduced risk of death [1]. Because of better survival and improved response of initial therapy with voriconazole, this agent is now considered the drug of choice for primary therapy of IA in most patients, including non-neutropenic patients, by the recent Clinical Practice Guidelines of the Infectious Diseases Society of America (IDSA) (strong recommendation, high-quality evidence) [5].
Isavuconazole is a new triazole agent that can be given once daily and offers a wider spectrum of antifungal activity than voriconazole, including activity against most Mucorales infections. In addition, the intravenous formulation does not include cyclodextrin, a nephrotoxic and hepatotoxic compound, which is included in the intravenous formulations of the other triazoles in order to increase solubility. Compared to voriconazole, isavuconazole also has the advantages of linear and predictable pharmacokinetics which is likely to obviate the need for therapeutic drug monitoring and fewer CYP enzyme-mediated drug interactions [66]. A large randomized, double-blind trial has demonstrated non-inferiority or isavuconazole versus voriconazole in terms of all-cause mortality when used as primary treatment for invasive fungal disease caused by Aspergillus species or other filamentous fungi, with a superior safety profile [16].
Another alternative for primary therapy is represented by amphotericin B that was historically considered the mainstay of treatment for IA before the introduction of voriconazole. Development of lipid formulations improved the poor tolerability associated with the deoxycholate formulation, but the optimal dosage remains unconfirmed [67].
All echinocandins have been shown to exert in vitro and in vivo activity against Aspergillus spp., but only caspofungin is approved for the treatment of IA in patients who are intolerant to first-line therapy [5].
Other azoles such as itraconazole or posaconazole are considered as second line agents for the treatment of IA, particularly in severely ill patients [5]. Use of these drugs in non-life-threatening infections where the patient has been already stabilized with a more potent agent has been described [64]. However, their applicability in non-neutropenic patients is limited because of scarce clinical experience, poor oral bioavailability and restricted access to the intravenous formulation [64]. A study comparing intravenous posaconazole versus voriconazole for primary therapy of invasive aspergillosis is ongoing (clinicaltrial.gov identifier NTC 01782131).
Although still not approved, the other two echinocandins (anidulafungin and micafungin) are used in clinical practice, particularly when non-neutropenic patients are involved. In breakthrough IA and in refractory disease, combination therapy (e.g., echinocandin plus voriconazole or liposomal amphotericin B) may be considered [5].
Adequate duration of antifungal therapy for IPA is an unresolved issue. Recent IDSA guidelines recommend treatment for IPA to be continued for a minimum of 6–12 weeks [5], depending on the clinical condition of the patients, as well as the extent of resolution of clinical disease. Different strategies including clinical evolution, serum biomarkers and CT scan should be considered for adequately monitoring therapeutic response for IA.
Some authors, including us, believe that emission tomography 18fluorodeoxygucose could be a new useful tool for monitoring response to IPA treatment [58, 68]. Although this technique is not specific, it may be helpful in monitoring clinical evolution of the patients, especially when biomarkers and CT scan are not enough.

Conclusion

There are several future challenges in the management of IA in non-neutropenic patients. Since new immunosuppressive regimens and ICU care are expected to continue increasing the incidence of populations at risk of IA, new criteria for diagnosis of IA in non-neutropenic patients are needed. We believe that there could be a potential role of FDG/PET-TC for treatment monitoring in IA. More pharmacokinetic/pharmacodynamics data on antifungal agents in non-neutropenic patients are needed to optimize drug exposure and to minimize adverse events, especially in patients with underlying severe disease and concomitant medications.

Acknowledgements

Funding

No funding or sponsorship was received for this study or publication of this article.

Authorship

All named authors meet the International Committee of Medical Journal Editors (ICMJE) criteria for authorship for this manuscript, take responsibility for the integrity of the work as a whole, and have given final approval for the version to be published.

Prior Presentation

The paper is based on the presentation “Challenges and solutions in the treatment of invasive mould infection in the ICU setting” presented by Matteo Bassetti on the Symposium “Challenges and new clinical treatment options for the management of invasive mould infections” sponsored by Basilea during the European Congress of Clinical Microbiology and Infectious Diseases (ECCMID) in Vienna on 24th April 2017.

Disclosures

Matteo Bassetti received research grants, consultation and/or speakers’ fees from Astellas, Pfizer, MSD, Gilead, Angelini, AstraZeneca, Bayer, Basilea, Gilead, Menarini, Novartis, Shionogi, Vifor, The Medicines company, Tetraphase, Achaogen, Paratek. Maddalena Peghin and Antonio Vena have nothing to disclose.

Compliance with Ethical Guidelines

This article is based on previously conducted studies and does not involve any studies of human or animal subjects performed by any of the authors.

Data Availability

Data sharing is not applicable to this article as no datasets were generated or analyzed during the current study.

Open Access

This article is distributed under the terms of the Creative Commons Attribution-NonCommercial 4.0 International License (http://​creativecommons.​org/​licenses/​by-nc/​4.​0/​), which permits any noncommercial use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (https://​creativecommons.​org/​licenses/​by/​4.​0), which permits use, duplication, 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 license, and indicate if changes were made.
Literatur
1.
Zurück zum Zitat Garcia-Vidal C, Peghin M, Cervera C, et al. Causes of death in a contemporary cohort of patients with invasive aspergillosis. PLoS One. 2015;10:e0120370.CrossRefPubMedPubMedCentral Garcia-Vidal C, Peghin M, Cervera C, et al. Causes of death in a contemporary cohort of patients with invasive aspergillosis. PLoS One. 2015;10:e0120370.CrossRefPubMedPubMedCentral
2.
Zurück zum Zitat Meersseman W, Vandecasteele SJ, Wilmer A, et al. Invasive aspergillosis in critically ill patients without malignancy. Am J Respir Crit Care Med. 2004;170:621–5.CrossRefPubMed Meersseman W, Vandecasteele SJ, Wilmer A, et al. Invasive aspergillosis in critically ill patients without malignancy. Am J Respir Crit Care Med. 2004;170:621–5.CrossRefPubMed
3.
Zurück zum Zitat Cornillet A, Camus C, Nimubona S, et al. Comparison of epidemiological, clinical, and biological features of invasive aspergillosis in neutropenic and non-neutropenic patients: a 6-year survey. Clin Infect Dis. 2006;43:577–84.CrossRefPubMed Cornillet A, Camus C, Nimubona S, et al. Comparison of epidemiological, clinical, and biological features of invasive aspergillosis in neutropenic and non-neutropenic patients: a 6-year survey. Clin Infect Dis. 2006;43:577–84.CrossRefPubMed
4.
5.
Zurück zum Zitat Patterson TF, Thompson GR 3rd, Denning DW, et al. Practice guidelines for the diagnosis and management of aspergillosis: 2016 update by the infectious diseases society of America. Clin Infect Dis. 2016;63:e1–60.CrossRefPubMedPubMedCentral Patterson TF, Thompson GR 3rd, Denning DW, et al. Practice guidelines for the diagnosis and management of aspergillosis: 2016 update by the infectious diseases society of America. Clin Infect Dis. 2016;63:e1–60.CrossRefPubMedPubMedCentral
6.
Zurück zum Zitat Meersseman W, Lagrou K, Maertens J, et al. Invasive aspergillosis in the intensive care unit. Clin Infect Dis. 2007;45:205–16.CrossRefPubMed Meersseman W, Lagrou K, Maertens J, et al. Invasive aspergillosis in the intensive care unit. Clin Infect Dis. 2007;45:205–16.CrossRefPubMed
7.
Zurück zum Zitat Baddley JW, Andes DR, Marr KA, et al. Factors associated with mortality in transplant patients with invasive aspergillosis. Clin Infect Dis. 2010;50:1559–67.CrossRefPubMedPubMedCentral Baddley JW, Andes DR, Marr KA, et al. Factors associated with mortality in transplant patients with invasive aspergillosis. Clin Infect Dis. 2010;50:1559–67.CrossRefPubMedPubMedCentral
8.
Zurück zum Zitat Koulenti D, Vogelaers D, Blot S. What’s new in invasive pulmonary aspergillosis in the critically ill. Intensive Care Med. 2014;40:723–6.CrossRefPubMed Koulenti D, Vogelaers D, Blot S. What’s new in invasive pulmonary aspergillosis in the critically ill. Intensive Care Med. 2014;40:723–6.CrossRefPubMed
9.
Zurück zum Zitat Taccone FS, Van den Abeele AM, Bulpa P, et al. Epidemiology of invasive aspergillosis in critically ill patients: clinical presentation, underlying conditions, and outcomes. Crit Care. 2015;19:7.CrossRefPubMedPubMedCentral Taccone FS, Van den Abeele AM, Bulpa P, et al. Epidemiology of invasive aspergillosis in critically ill patients: clinical presentation, underlying conditions, and outcomes. Crit Care. 2015;19:7.CrossRefPubMedPubMedCentral
10.
Zurück zum Zitat Segal BH, Romani LR. Invasive aspergillosis in chronic granulomatous disease. Med Mycol. 2009;47(Suppl 1):S282–90.CrossRefPubMed Segal BH, Romani LR. Invasive aspergillosis in chronic granulomatous disease. Med Mycol. 2009;47(Suppl 1):S282–90.CrossRefPubMed
11.
Zurück zum Zitat De Pauw B, Walsh TJ, Donnelly JP, et al. Revised definitions of invasive fungal disease from the European Organization for Research and Treatment of Cancer/Invasive Fungal Infections Cooperative Group and the National Institute of Allergy and Infectious Diseases Mycoses Study Group (EORTC/MSG) Consensus Group. Clin Infect Dis. 2008;46:1813–21.CrossRefPubMedPubMedCentral De Pauw B, Walsh TJ, Donnelly JP, et al. Revised definitions of invasive fungal disease from the European Organization for Research and Treatment of Cancer/Invasive Fungal Infections Cooperative Group and the National Institute of Allergy and Infectious Diseases Mycoses Study Group (EORTC/MSG) Consensus Group. Clin Infect Dis. 2008;46:1813–21.CrossRefPubMedPubMedCentral
12.
Zurück zum Zitat Blot SI, Taccone FS, Van den Abeele AM, et al. A clinical algorithm to diagnose invasive pulmonary aspergillosis in critically ill patients. Am J Respir Crit Care Med. 2012;186:56–64.CrossRefPubMed Blot SI, Taccone FS, Van den Abeele AM, et al. A clinical algorithm to diagnose invasive pulmonary aspergillosis in critically ill patients. Am J Respir Crit Care Med. 2012;186:56–64.CrossRefPubMed
13.
Zurück zum Zitat Munoz P, Vena A, Ceron I, et al. Invasive pulmonary aspergillosis in heart transplant recipients: two radiologic patterns with a different prognosis. J Heart Lung Transplant. 2014;33:1034–40.CrossRefPubMed Munoz P, Vena A, Ceron I, et al. Invasive pulmonary aspergillosis in heart transplant recipients: two radiologic patterns with a different prognosis. J Heart Lung Transplant. 2014;33:1034–40.CrossRefPubMed
14.
Zurück zum Zitat Prattes J, Hoenigl M, Krause R, et al. Invasive aspergillosis in patients with underlying liver cirrhosis: a prospective cohort study. Med Mycol. 2017;55(8):803–12.CrossRefPubMed Prattes J, Hoenigl M, Krause R, et al. Invasive aspergillosis in patients with underlying liver cirrhosis: a prospective cohort study. Med Mycol. 2017;55(8):803–12.CrossRefPubMed
15.
Zurück zum Zitat Olaechea Astigarraga PM, Alvarez Lerma F. Zaldibar Enriquez E [Invasive pulmonary aspergillosis in the non-neutropenic critical patient: future challenges]. Med Intensiva. 2006;30:386–91.CrossRefPubMed Olaechea Astigarraga PM, Alvarez Lerma F. Zaldibar Enriquez E [Invasive pulmonary aspergillosis in the non-neutropenic critical patient: future challenges]. Med Intensiva. 2006;30:386–91.CrossRefPubMed
16.
Zurück zum Zitat Maertens JA, Raad II, Marr KA, et al. Isavuconazole versus voriconazole for primary treatment of invasive mould disease caused by Aspergillus and other filamentous fungi (SECURE): a phase 3, randomised-controlled, non-inferiority trial. Lancet. 2016;387:760–9.CrossRefPubMed Maertens JA, Raad II, Marr KA, et al. Isavuconazole versus voriconazole for primary treatment of invasive mould disease caused by Aspergillus and other filamentous fungi (SECURE): a phase 3, randomised-controlled, non-inferiority trial. Lancet. 2016;387:760–9.CrossRefPubMed
17.
Zurück zum Zitat Marr KA, Schlamm HT, Herbrecht R, et al. Combination antifungal therapy for invasive aspergillosis: a randomized trial. Ann Intern Med. 2015;162:81–9.CrossRefPubMed Marr KA, Schlamm HT, Herbrecht R, et al. Combination antifungal therapy for invasive aspergillosis: a randomized trial. Ann Intern Med. 2015;162:81–9.CrossRefPubMed
18.
Zurück zum Zitat Bassetti M, Bouza E. Invasive mould infections in the ICU setting: complexities and solutions. J Antimicrob Chemother. 2017;72:i39–47.CrossRefPubMed Bassetti M, Bouza E. Invasive mould infections in the ICU setting: complexities and solutions. J Antimicrob Chemother. 2017;72:i39–47.CrossRefPubMed
19.
Zurück zum Zitat Bretagne S, Marmorat-Khuong A, Kuentz M, et al. Serum Aspergillus galactomannan antigen testing by sandwich ELISA: practical use in neutropenic patients. J Infect. 1997;35:7–15.CrossRefPubMed Bretagne S, Marmorat-Khuong A, Kuentz M, et al. Serum Aspergillus galactomannan antigen testing by sandwich ELISA: practical use in neutropenic patients. J Infect. 1997;35:7–15.CrossRefPubMed
20.
Zurück zum Zitat Maertens J, Verhaegen J, Lagrou K, et al. Screening for circulating galactomannan as a noninvasive diagnostic tool for invasive aspergillosis in prolonged neutropenic patients and stem cell transplantation recipients: a prospective validation. Blood. 2001;97:1604–10.CrossRefPubMed Maertens J, Verhaegen J, Lagrou K, et al. Screening for circulating galactomannan as a noninvasive diagnostic tool for invasive aspergillosis in prolonged neutropenic patients and stem cell transplantation recipients: a prospective validation. Blood. 2001;97:1604–10.CrossRefPubMed
21.
Zurück zum Zitat Pfeiffer CD, Fine JP, Safdar N. Diagnosis of invasive aspergillosis using a galactomannan assay: a meta-analysis. Clin Infect Dis. 2006;42:1417–27.CrossRefPubMed Pfeiffer CD, Fine JP, Safdar N. Diagnosis of invasive aspergillosis using a galactomannan assay: a meta-analysis. Clin Infect Dis. 2006;42:1417–27.CrossRefPubMed
22.
Zurück zum Zitat Zhou W, Li H, Zhang Y, et al. Diagnostic value of galactomannan antigen test in serum and bronchoalveolar lavage fluid samples from patients with nonneutropenic invasive pulmonary aspergillosis. J Clin Microbiol. 2017;55:2153–61.CrossRefPubMedPubMedCentral Zhou W, Li H, Zhang Y, et al. Diagnostic value of galactomannan antigen test in serum and bronchoalveolar lavage fluid samples from patients with nonneutropenic invasive pulmonary aspergillosis. J Clin Microbiol. 2017;55:2153–61.CrossRefPubMedPubMedCentral
23.
Zurück zum Zitat Guinea J, Bouza E. Current challenges in the microbiological diagnosis of invasive aspergillosis. Mycopathologia. 2014;178:403–16.CrossRefPubMed Guinea J, Bouza E. Current challenges in the microbiological diagnosis of invasive aspergillosis. Mycopathologia. 2014;178:403–16.CrossRefPubMed
24.
Zurück zum Zitat Mikulska M, Furfaro E, Viscoli C. Non-cultural methods for the diagnosis of invasive fungal disease. Expert Rev Anti Infect Ther. 2015;13:103–17.CrossRefPubMed Mikulska M, Furfaro E, Viscoli C. Non-cultural methods for the diagnosis of invasive fungal disease. Expert Rev Anti Infect Ther. 2015;13:103–17.CrossRefPubMed
25.
Zurück zum Zitat Oz Y, Aslan M, Aksit F, et al. The effect of clinical characteristics on the performance of galactomannan and PCR for the diagnosis of invasive aspergillosis in febrile neutropenic patients. Mycoses. 2016;59:86–92.CrossRefPubMed Oz Y, Aslan M, Aksit F, et al. The effect of clinical characteristics on the performance of galactomannan and PCR for the diagnosis of invasive aspergillosis in febrile neutropenic patients. Mycoses. 2016;59:86–92.CrossRefPubMed
26.
Zurück zum Zitat Kovanda LL, Kolamunnage-Dona R, Neely M, et al. Pharmacodynamics of isavuconazole for invasive mold disease: role of galactomannan for real-time monitoring of therapeutic response. Clin Infect Dis. 2017;64:1557–63.CrossRefPubMedPubMedCentral Kovanda LL, Kolamunnage-Dona R, Neely M, et al. Pharmacodynamics of isavuconazole for invasive mold disease: role of galactomannan for real-time monitoring of therapeutic response. Clin Infect Dis. 2017;64:1557–63.CrossRefPubMedPubMedCentral
27.
Zurück zum Zitat Neofytos D, Railkar R, Mullane KM, et al. Correlation between circulating fungal biomarkers and clinical outcome in invasive aspergillosis. PLoS ONE. 2015;10:e0129022.CrossRefPubMedPubMedCentral Neofytos D, Railkar R, Mullane KM, et al. Correlation between circulating fungal biomarkers and clinical outcome in invasive aspergillosis. PLoS ONE. 2015;10:e0129022.CrossRefPubMedPubMedCentral
28.
Zurück zum Zitat Chai LY, Kullberg BJ, Johnson EM, et al. Early serum galactomannan trend as a predictor of outcome of invasive aspergillosis. J Clin Microbiol. 2012;50:2330–6.CrossRefPubMedPubMedCentral Chai LY, Kullberg BJ, Johnson EM, et al. Early serum galactomannan trend as a predictor of outcome of invasive aspergillosis. J Clin Microbiol. 2012;50:2330–6.CrossRefPubMedPubMedCentral
29.
Zurück zum Zitat Russo A, Giuliano S, Vena A, et al. Predictors of mortality in non-neutropenic patients with invasive pulmonary aspergillosis: does galactomannan have a role? Diagn Microbiol Infect Dis. 2014;80:83–6.CrossRefPubMed Russo A, Giuliano S, Vena A, et al. Predictors of mortality in non-neutropenic patients with invasive pulmonary aspergillosis: does galactomannan have a role? Diagn Microbiol Infect Dis. 2014;80:83–6.CrossRefPubMed
30.
Zurück zum Zitat Karageorgopoulos DE, Vouloumanou EK, Ntziora F, et al. beta-d-glucan assay for the diagnosis of invasive fungal infections: a meta-analysis. Clin Infect Dis. 2011;52:750–70.CrossRefPubMed Karageorgopoulos DE, Vouloumanou EK, Ntziora F, et al. beta-d-glucan assay for the diagnosis of invasive fungal infections: a meta-analysis. Clin Infect Dis. 2011;52:750–70.CrossRefPubMed
31.
Zurück zum Zitat Lu Y, Chen YQ, Guo YL, et al. Diagnosis of invasive fungal disease using serum (1– > 3)-beta-d-glucan: a bivariate meta-analysis. Intern Med. 2011;50:2783–91.CrossRefPubMed Lu Y, Chen YQ, Guo YL, et al. Diagnosis of invasive fungal disease using serum (1– > 3)-beta-d-glucan: a bivariate meta-analysis. Intern Med. 2011;50:2783–91.CrossRefPubMed
32.
Zurück zum Zitat Lamoth F, Cruciani M, Mengoli C, et al. Beta-Glucan antigenemia assay for the diagnosis of invasive fungal infections in patients with hematological malignancies: a systematic review and meta-analysis of cohort studies from the Third European Conference on Infections in Leukemia (ECIL-3). Clin Infect Dis. 2012;54:633–43.CrossRefPubMed Lamoth F, Cruciani M, Mengoli C, et al. Beta-Glucan antigenemia assay for the diagnosis of invasive fungal infections in patients with hematological malignancies: a systematic review and meta-analysis of cohort studies from the Third European Conference on Infections in Leukemia (ECIL-3). Clin Infect Dis. 2012;54:633–43.CrossRefPubMed
33.
Zurück zum Zitat Onishi A, Sugiyama D, Kogata Y, et al. Diagnostic accuracy of serum 1,3-beta-d-glucan for pneumocystis jiroveci pneumonia, invasive candidiasis, and invasive aspergillosis: systematic review and meta-analysis. J Clin Microbiol. 2012;50:7–15.CrossRefPubMedPubMedCentral Onishi A, Sugiyama D, Kogata Y, et al. Diagnostic accuracy of serum 1,3-beta-d-glucan for pneumocystis jiroveci pneumonia, invasive candidiasis, and invasive aspergillosis: systematic review and meta-analysis. J Clin Microbiol. 2012;50:7–15.CrossRefPubMedPubMedCentral
34.
Zurück zum Zitat Maertens JA, Blennow O, Duarte RF, et al. The current management landscape: aspergillosis. J Antimicrob Chemother. 2016;71:23–9.CrossRef Maertens JA, Blennow O, Duarte RF, et al. The current management landscape: aspergillosis. J Antimicrob Chemother. 2016;71:23–9.CrossRef
35.
Zurück zum Zitat Theel ES, Jespersen DJ, Iqbal S, et al. Detection of (1, 3)-beta-d-glucan in bronchoalveolar lavage and serum samples collected from immunocompromised hosts. Mycopathologia. 2013;175:33–41.CrossRefPubMed Theel ES, Jespersen DJ, Iqbal S, et al. Detection of (1, 3)-beta-d-glucan in bronchoalveolar lavage and serum samples collected from immunocompromised hosts. Mycopathologia. 2013;175:33–41.CrossRefPubMed
37.
Zurück zum Zitat Arvanitis M, Ziakas PD, Zacharioudakis IM, et al. PCR in diagnosis of invasive aspergillosis: a meta-analysis of diagnostic performance. J Clin Microbiol. 2014;52:3731–42.CrossRefPubMedPubMedCentral Arvanitis M, Ziakas PD, Zacharioudakis IM, et al. PCR in diagnosis of invasive aspergillosis: a meta-analysis of diagnostic performance. J Clin Microbiol. 2014;52:3731–42.CrossRefPubMedPubMedCentral
38.
Zurück zum Zitat White PL, Barnes RA, Springer J, et al. Clinical performance of Aspergillus PCR for testing serum and plasma: a study by the European Aspergillus PCR initiative. J Clin Microbiol. 2015;53:2832–7.CrossRefPubMedPubMedCentral White PL, Barnes RA, Springer J, et al. Clinical performance of Aspergillus PCR for testing serum and plasma: a study by the European Aspergillus PCR initiative. J Clin Microbiol. 2015;53:2832–7.CrossRefPubMedPubMedCentral
39.
Zurück zum Zitat Loeffler J, Mengoli C, Springer J, et al. Analytical comparison of in vitro-spiked human serum and plasma for PCR-based detection of Aspergillus fumigatus DNA: a study by the European Aspergillus PCR initiative. J Clin Microbiol. 2015;53:2838–45.CrossRefPubMedPubMedCentral Loeffler J, Mengoli C, Springer J, et al. Analytical comparison of in vitro-spiked human serum and plasma for PCR-based detection of Aspergillus fumigatus DNA: a study by the European Aspergillus PCR initiative. J Clin Microbiol. 2015;53:2838–45.CrossRefPubMedPubMedCentral
40.
Zurück zum Zitat Springer J, Morton CO, Perry M, et al. Multicenter comparison of serum and whole-blood specimens for detection of Aspergillus DNA in high-risk hematological patients. J Clin Microbiol. 2013;51:1445–50.CrossRefPubMedPubMedCentral Springer J, Morton CO, Perry M, et al. Multicenter comparison of serum and whole-blood specimens for detection of Aspergillus DNA in high-risk hematological patients. J Clin Microbiol. 2013;51:1445–50.CrossRefPubMedPubMedCentral
41.
Zurück zum Zitat Springer J, White PL, Hamilton S, et al. Comparison of performance characteristics of Aspergillus PCR in testing a range of blood-based samples in accordance with international methodological recommendations. J Clin Microbiol. 2016;54:705–11.CrossRefPubMedPubMedCentral Springer J, White PL, Hamilton S, et al. Comparison of performance characteristics of Aspergillus PCR in testing a range of blood-based samples in accordance with international methodological recommendations. J Clin Microbiol. 2016;54:705–11.CrossRefPubMedPubMedCentral
42.
Zurück zum Zitat Mengoli C, Cruciani M, Barnes RA, et al. Use of PCR for diagnosis of invasive aspergillosis: systematic review and meta-analysis. Lancet Infect Dis. 2009;9:89–96.CrossRefPubMed Mengoli C, Cruciani M, Barnes RA, et al. Use of PCR for diagnosis of invasive aspergillosis: systematic review and meta-analysis. Lancet Infect Dis. 2009;9:89–96.CrossRefPubMed
43.
Zurück zum Zitat Imbert S, Gauthier L, Joly I, et al. Aspergillus PCR in serum for the diagnosis, follow-up and prognosis of invasive aspergillosis in neutropenic and nonneutropenic patients. Clin Microbiol Infect. 2016;22(562):e1–8. Imbert S, Gauthier L, Joly I, et al. Aspergillus PCR in serum for the diagnosis, follow-up and prognosis of invasive aspergillosis in neutropenic and nonneutropenic patients. Clin Microbiol Infect. 2016;22(562):e1–8.
44.
Zurück zum Zitat Chong GL, van de Sande WW, Dingemans GJ, et al. Validation of a new Aspergillus real-time PCR assay for direct detection of Aspergillus and azole resistance of Aspergillus fumigatus on bronchoalveolar lavage fluid. J Clin Microbiol. 2015;53:868–74.CrossRefPubMedPubMedCentral Chong GL, van de Sande WW, Dingemans GJ, et al. Validation of a new Aspergillus real-time PCR assay for direct detection of Aspergillus and azole resistance of Aspergillus fumigatus on bronchoalveolar lavage fluid. J Clin Microbiol. 2015;53:868–74.CrossRefPubMedPubMedCentral
45.
Zurück zum Zitat Held J, Schmidt T, Thornton CR, et al. Comparison of a novel Aspergillus lateral-flow device and the Platelia(R) galactomannan assay for the diagnosis of invasive aspergillosis following haematopoietic stem cell transplantation. Infection. 2013;41:1163–9.CrossRefPubMed Held J, Schmidt T, Thornton CR, et al. Comparison of a novel Aspergillus lateral-flow device and the Platelia(R) galactomannan assay for the diagnosis of invasive aspergillosis following haematopoietic stem cell transplantation. Infection. 2013;41:1163–9.CrossRefPubMed
46.
Zurück zum Zitat Willinger B, Lackner M, Lass-Florl C, et al. Bronchoalveolar lavage lateral-flow device test for invasive pulmonary aspergillosis in solid organ transplant patients: a semiprospective multicenter study. Transplantation. 2014;98:898–902.CrossRefPubMed Willinger B, Lackner M, Lass-Florl C, et al. Bronchoalveolar lavage lateral-flow device test for invasive pulmonary aspergillosis in solid organ transplant patients: a semiprospective multicenter study. Transplantation. 2014;98:898–902.CrossRefPubMed
47.
Zurück zum Zitat Prattes J, Flick H, Pruller F, et al. Novel tests for diagnosis of invasive aspergillosis in patients with underlying respiratory diseases. Am J Respir Crit Care Med. 2014;190:922–9.CrossRefPubMed Prattes J, Flick H, Pruller F, et al. Novel tests for diagnosis of invasive aspergillosis in patients with underlying respiratory diseases. Am J Respir Crit Care Med. 2014;190:922–9.CrossRefPubMed
48.
Zurück zum Zitat Eigl S, Prattes J, Lackner M, et al. Multicenter evaluation of a lateral-flow device test for diagnosing invasive pulmonary aspergillosis in ICU patients. Crit Care. 2015;19:178.CrossRefPubMedPubMedCentral Eigl S, Prattes J, Lackner M, et al. Multicenter evaluation of a lateral-flow device test for diagnosing invasive pulmonary aspergillosis in ICU patients. Crit Care. 2015;19:178.CrossRefPubMedPubMedCentral
49.
Zurück zum Zitat de Heer K, van der Schee MP, Zwinderman K, et al. Electronic nose technology for detection of invasive pulmonary aspergillosis in prolonged chemotherapy-induced neutropenia: a proof-of-principle study. J Clin Microbiol. 2013;51:1490–5.CrossRefPubMedPubMedCentral de Heer K, van der Schee MP, Zwinderman K, et al. Electronic nose technology for detection of invasive pulmonary aspergillosis in prolonged chemotherapy-induced neutropenia: a proof-of-principle study. J Clin Microbiol. 2013;51:1490–5.CrossRefPubMedPubMedCentral
50.
Zurück zum Zitat Koo S, Thomas HR, Daniels SD, et al. A breath fungal secondary metabolite signature to diagnose invasive aspergillosis. Clin Infect Dis. 2014;59:1733–40.CrossRefPubMedPubMedCentral Koo S, Thomas HR, Daniels SD, et al. A breath fungal secondary metabolite signature to diagnose invasive aspergillosis. Clin Infect Dis. 2014;59:1733–40.CrossRefPubMedPubMedCentral
51.
Zurück zum Zitat Domingo MP, Colmenarejo C, Martinez-Lostao L, et al. Bis(methyl)gliotoxin proves to be a more stable and reliable marker for invasive aspergillosis than gliotoxin and suitable for use in diagnosis. Diagn Microbiol Infect Dis. 2012;73:57–64.CrossRefPubMed Domingo MP, Colmenarejo C, Martinez-Lostao L, et al. Bis(methyl)gliotoxin proves to be a more stable and reliable marker for invasive aspergillosis than gliotoxin and suitable for use in diagnosis. Diagn Microbiol Infect Dis. 2012;73:57–64.CrossRefPubMed
52.
Zurück zum Zitat Vidal-Garcia M, Domingo MP, De Rueda B, et al. Clinical validity of bis(methylthio)gliotoxin for the diagnosis of invasive aspergillosis. Appl Microbiol Biotechnol. 2016;100:2327–34.CrossRefPubMed Vidal-Garcia M, Domingo MP, De Rueda B, et al. Clinical validity of bis(methylthio)gliotoxin for the diagnosis of invasive aspergillosis. Appl Microbiol Biotechnol. 2016;100:2327–34.CrossRefPubMed
53.
Zurück zum Zitat Dai Z, Zhao H, Cai S, et al. Invasive pulmonary aspergillosis in non-neutropenic patients with and without underlying disease: a single-centre retrospective analysis of 52 subjects. Respirology. 2013;18:323–31.CrossRefPubMed Dai Z, Zhao H, Cai S, et al. Invasive pulmonary aspergillosis in non-neutropenic patients with and without underlying disease: a single-centre retrospective analysis of 52 subjects. Respirology. 2013;18:323–31.CrossRefPubMed
54.
Zurück zum Zitat Henzler C, Henzler T, Buchheidt D, et al. Diagnostic performance of contrast enhanced pulmonary computed tomography angiography for the detection of angioinvasive pulmonary aspergillosis in immunocompromised patients. Sci Rep. 2017;7:4483.CrossRefPubMedPubMedCentral Henzler C, Henzler T, Buchheidt D, et al. Diagnostic performance of contrast enhanced pulmonary computed tomography angiography for the detection of angioinvasive pulmonary aspergillosis in immunocompromised patients. Sci Rep. 2017;7:4483.CrossRefPubMedPubMedCentral
55.
Zurück zum Zitat Bassetti M, Carnelutti A, Muser D, et al. 18F-fluorodeoxyglucose positron emission tomography and infectious diseases: current applications and future perspectives. Curr Opin Infect Dis. 2017;30:192–200.CrossRefPubMed Bassetti M, Carnelutti A, Muser D, et al. 18F-fluorodeoxyglucose positron emission tomography and infectious diseases: current applications and future perspectives. Curr Opin Infect Dis. 2017;30:192–200.CrossRefPubMed
56.
Zurück zum Zitat Kim JY, Yoo JW, Oh M, et al. (18)F-fluoro-2-deoxy-d-glucose positron emission tomography/computed tomography findings are different between invasive and noninvasive pulmonary aspergillosis. J Comput Assist Tomogr. 2013;37:596–601.CrossRefPubMed Kim JY, Yoo JW, Oh M, et al. (18)F-fluoro-2-deoxy-d-glucose positron emission tomography/computed tomography findings are different between invasive and noninvasive pulmonary aspergillosis. J Comput Assist Tomogr. 2013;37:596–601.CrossRefPubMed
57.
Zurück zum Zitat Hot A, Maunoury C, Poiree S, et al. Diagnostic contribution of positron emission tomography with [18F]fluorodeoxyglucose for invasive fungal infections. Clin Microbiol Infect. 2011;17:409–17.CrossRefPubMed Hot A, Maunoury C, Poiree S, et al. Diagnostic contribution of positron emission tomography with [18F]fluorodeoxyglucose for invasive fungal infections. Clin Microbiol Infect. 2011;17:409–17.CrossRefPubMed
58.
Zurück zum Zitat Rolle AM, Hasenberg M, Thornton CR, et al. ImmunoPET/MR imaging allows specific detection of Aspergillus fumigatus lung infection in vivo. Proc Natl Acad Sci USA. 2016;113:E1026–33.CrossRefPubMedPubMedCentral Rolle AM, Hasenberg M, Thornton CR, et al. ImmunoPET/MR imaging allows specific detection of Aspergillus fumigatus lung infection in vivo. Proc Natl Acad Sci USA. 2016;113:E1026–33.CrossRefPubMedPubMedCentral
59.
Zurück zum Zitat Marr KA. Empirical antifungal therapy—new options, new tradeoffs. N Engl J Med. 2002;346:278–80.CrossRefPubMed Marr KA. Empirical antifungal therapy—new options, new tradeoffs. N Engl J Med. 2002;346:278–80.CrossRefPubMed
60.
Zurück zum Zitat Cornely OA, Meems L, Herbrecht R, et al. Randomised, multicentre trial of micafungin vs. an institutional standard regimen for salvage treatment of invasive aspergillosis. Mycoses. 2015;58:58–64.CrossRefPubMed Cornely OA, Meems L, Herbrecht R, et al. Randomised, multicentre trial of micafungin vs. an institutional standard regimen for salvage treatment of invasive aspergillosis. Mycoses. 2015;58:58–64.CrossRefPubMed
61.
Zurück zum Zitat Cattaneo C, Monte S, Algarotti A, et al. A randomized comparison of caspofungin versus antifungal prophylaxis according to investigator policy in acute leukaemia patients undergoing induction chemotherapy (PROFIL-C study). J Antimicrob Chemother. 2011;66:2140–5.CrossRefPubMed Cattaneo C, Monte S, Algarotti A, et al. A randomized comparison of caspofungin versus antifungal prophylaxis according to investigator policy in acute leukaemia patients undergoing induction chemotherapy (PROFIL-C study). J Antimicrob Chemother. 2011;66:2140–5.CrossRefPubMed
62.
Zurück zum Zitat Hiemenz JW, Raad II, Maertens JA, et al. Efficacy of caspofungin as salvage therapy for invasive aspergillosis compared to standard therapy in a historical cohort. Eur J Clin Microbiol Infect Dis. 2010;29:1387–94.CrossRefPubMed Hiemenz JW, Raad II, Maertens JA, et al. Efficacy of caspofungin as salvage therapy for invasive aspergillosis compared to standard therapy in a historical cohort. Eur J Clin Microbiol Infect Dis. 2010;29:1387–94.CrossRefPubMed
63.
Zurück zum Zitat Herbrecht R, Denning DW, Patterson TF, 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, et al. Voriconazole versus amphotericin B for primary therapy of invasive aspergillosis. N Engl J Med. 2002;347:408–15.CrossRefPubMed
64.
Zurück zum Zitat Ledoux MP, Toussaint E, Denis J, et al. New pharmacological opportunities for the treatment of invasive mould diseases. J Antimicrob Chemother. 2017;72:i48–58.CrossRefPubMed Ledoux MP, Toussaint E, Denis J, et al. New pharmacological opportunities for the treatment of invasive mould diseases. J Antimicrob Chemother. 2017;72:i48–58.CrossRefPubMed
65.
Zurück zum Zitat Baddley JW, Stephens JM, Ji X, et al. Aspergillosis in intensive care unit (ICU) patients: epidemiology and economic outcomes. BMC Infect Dis. 2013;13:29.CrossRefPubMedPubMedCentral Baddley JW, Stephens JM, Ji X, et al. Aspergillosis in intensive care unit (ICU) patients: epidemiology and economic outcomes. BMC Infect Dis. 2013;13:29.CrossRefPubMedPubMedCentral
66.
Zurück zum Zitat Miceli MH, Kauffman CA. Isavuconazole: a new broad-spectrum triazole antifungal agent. Clin Infect Dis. 2015;61:1558–65.CrossRefPubMed Miceli MH, Kauffman CA. Isavuconazole: a new broad-spectrum triazole antifungal agent. Clin Infect Dis. 2015;61:1558–65.CrossRefPubMed
67.
Zurück zum Zitat Cornely OA, Maertens J, Bresnik M, et al. Liposomal amphotericin B as initial therapy for invasive mold infection: a randomized trial comparing a high-loading dose regimen with standard dosing (AmBiLoad trial). Clin Infect Dis. 2007;44:1289–97.CrossRefPubMed Cornely OA, Maertens J, Bresnik M, et al. Liposomal amphotericin B as initial therapy for invasive mold infection: a randomized trial comparing a high-loading dose regimen with standard dosing (AmBiLoad trial). Clin Infect Dis. 2007;44:1289–97.CrossRefPubMed
68.
Zurück zum Zitat Altini C, Niccoli Asabella A, Ferrari C, et al. (18)F-FDG PET/CT contribution to diagnosis and treatment response of rhino-orbital-cerebral mucormycosis. Hell J Nucl Med. 2015;18:68–70.PubMed Altini C, Niccoli Asabella A, Ferrari C, et al. (18)F-FDG PET/CT contribution to diagnosis and treatment response of rhino-orbital-cerebral mucormycosis. Hell J Nucl Med. 2015;18:68–70.PubMed
Metadaten
Titel
Challenges and Solution of Invasive Aspergillosis in Non-neutropenic Patients: A Review
verfasst von
Matteo Bassetti
Maddalena Peghin
Antonio Vena
Publikationsdatum
22.12.2017
Verlag
Springer Healthcare
Erschienen in
Infectious Diseases and Therapy / Ausgabe 1/2018
Print ISSN: 2193-8229
Elektronische ISSN: 2193-6382
DOI
https://doi.org/10.1007/s40121-017-0183-9

Weitere Artikel der Ausgabe 1/2018

Infectious Diseases and Therapy 1/2018 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Update Innere Medizin

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