Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2012 Apr 15.
Published in final edited form as: Curr Opin Pulm Med. 2010 May;16(3):242–250. doi: 10.1097/MCP.0b013e328337d6de

Pulmonary Aspergillosis: clinical presentation, diagnostic tests, management and complications

Rami Sherif 1, Brahm H Segal 1
PMCID: PMC3326383  NIHMSID: NIHMS213349  PMID: 20375786

Abstract

Purpose of Review

When functioning properly, the immune system recognizes inhaled fungi and controls their growth, while avoiding injurious inflammation and allergy. “Aspergillosis” represents a spectrum of clinical diseases resulting from impaired or excessive immune responses. Invasive aspergillosis is principally disease of severely immunocompromised patients, while allergic forms of aspergillosis result from an excessive inflammatory response to hyphae colonizing the sinopulmonary tract. We will review insights gained in host defense against Aspergillus species and the immunopathogenesis of Aspergillus-related diseases as well as important advances made in fungal diagnostics and antifungal therapy.

Recent Findings

Important advances have been made in diagnosis of invasive aspergillosis and in antifungal agents. Voriconazole was superior to amphotericin B deoxycholate as primary therapy for invasive aspergillosis. There is significant interest in combination antifungal therapy for invasive aspergillosis. Fungal genomics offer a powerful opportunity to gain knowledge about fungal virulence factors that can be targets for drug development. In addition, new insights have been gained regarding host defense against Aspergillus species that may be exploited therapeutically.

Summary

We have gained substantial knowledge regarding how the immune system recognizes inhaled fungi and calibrates the inflammatory response. There has also been substantial progress in tools to diagnose aspergillosis and in antifungal therapeutics. Future progress will likely involve the development of more refined diagnostic tools, new classes of antifungal agents, and greater knowledge of pathogen and host factors that predispose to aspergillosis.

Keywords: Aspergillosis, Allergy, Immunity, Immunocompromised, Antifungal

Introduction

Aspergillus species are ancient lineages [1] that are ubiquitous in the environment and grow independently of an animal host. Certain pathways used by Aspergillus species as saprophytes in the environment (e.g., for nutrient acquisition or regulation of pH) may also enhance its virulence as a human pathogen in a susceptible host [2]. The evolution from the onset of primitive immune systems such as those in insects [3] to the complex innate and antigen-driven immune system that exist in humans and other mammals occurred in the context of continual exposure to filamentous fungi. Therefore, our immune system requires the ability to recognize inhaled mould spores and to control their growth, but also to avoid excessive inflammation [4].

Alveolar macrophages (AM) constitute the first line of phagocytic host defense against inhaled conidia [5]. Following germination, neutrophils are the dominant host defense arm against hyphae, the tissue invasive form of moulds [5]. Pathogen recognitions receptors (PRRs) recognize specific fungal cell wall motifs displayed during the conidial and hyphal stage and produce cytokines and chemokines that stimulate neutrophil recruitment and subsequent antigen-specific immunity.

There are several classes of innate PRRs that recognize fungal motifs. Examples include toll-like receptors (TLRs,) dectin-1, pentraxin-3, SP-A, SP-D, and mannose-binding lectin. Fungal cell wall beta-glucans act as a trigger for the induction of inflammatory responses in macrophages through their time-dependent exposure on the surface of germinating conidia [68]. Dectin-1 and TLRs permit macrophages to distinguish between A. fumigatus conidia and hyphae. In A. fumigatus, a surface hydrophobic protein, RodA, is covalently bound to the conidial cell wall and prevents innate immune and T-helper responses [9]. Stage-specific activation of innate immunity via coordinated ligation of specific PRRs likely calibrates the immune response to inhaled fungi to contain fungal growth while restraining excessive inflammation.

Activation of dectin-1 can lead to NADPH oxidase activation [10]. The neutrophil NADPH oxidase is essential in host defense against aspergillosis. Chronic granulomatous disease (CGD), an inherited disorder of the NADPH oxidase, is associated with recurrent bacterial and fungal diseases, with invasive aspergillosis being a major cause of mortality [11]. NADPH oxidase activation results in conversion of oxygen to superoxide anion that is in turn coupled to activation of preformed antimicrobial proteases sequestered in the primary granules of neutrophils [12].

Dectin-1 ligation can also lead to production of proinflammatory cytokines, such as IL-17 [13]. IL-17A influences antimicrobial host defense, but also promotes inflammatory pathology in autoimmune disease [14]. IL-17A signals through IL-17 receptor A (IL-17R), and stimulates production of G-CSF, GM-CSF, TNF-α, and chemokines that regulate myelopoiesis and neutrophil recruitment to inflammatory sites. There is debate regarding in what circumstances IL-17 may enhance versus diminish antifungal immunity [15]. In humans, mutations leading to defective dectin-1 signaling are associated with chronic mucocutaneous candidiasis [16,17].

Dectin-1-deficient mice have increased susceptibility to aspergillosis in association with impaired NADPH oxidase activity and impaired production of proinflammatory cytokines and chemokines [18]. IL-17 production in the lungs after A. fumigatus challenge was Dectin-1 dependent, and neutralization of IL-17 impaired A. fumigatus clearance. Interestingly, NADPH oxidase restrains IL-17 responses and augments regulatory T-cell activity via tryptophan catabolism in mice in experimental aspergillosis [19]. These findings in mice illustrate the complex interactions mediated by pathogen recognition receptors that sense specific Aspergillus motifs and control fungal growth while calibrating the immune response to avert tissue injury.

Aspergillus species cause disease only in a small proportion of persons with specific host factors – a reflection of how well the immune system functions to control fungal growth and restrain excessive inflammation. Invasive aspergillosis is typically, although not exclusively, a disease of the highly immunocompromised. Allergic forms of aspergillosis, notably allergic bronchopulmonary aspergillosis (ABPA), result from a poorly controlled allergic response to hyphae colonizing human airways. Seen in this light, Aspergillus-related diseases result principally from disorders of immunity, and immune responses govern the clinical and pathological manifestations of aspergillosis.

Invasive aspergillosis: Host factors

Acute invasive aspergillosis is a devastating opportunistic infection in the severely immunocompromised (Table 1). Patients at risk for invasive aspergillosis include those with prolonged neutropenia (e.g., following cytotoxic regimens for acute leukemia), hematopoietic stem cell transplant (HSCT) recipients, solid organ transplant recipients (particularly lung transplant recipients), advanced AIDS and CGD [4,27]. Mortality from invasive aspergillosis has increased by several-fold in the 1980s and 1990s in the U.S. and Europe based on unselected autopsy reports [28,29] -- a reflection of more patients undergoing treatment for hematologic malignancies and allogeneic HSCT.

Table 1.

Predisposing host factors and immunopathogenesis of invasive, saprophytic, and allergic bronchopulmonary aspergillosis

Patient populations Predisposing host factors Clinical and histological features
Acute leukemia, myelodysplastic syndrome, aplastic anemia, other causes of marrow failure Neutropenia Hyphal angioinvasion with vascular thrombosis and tissue infarction; scant inflammatory response; may evolve to cavitation
Allogeneic HSCT after neutrophil recovery Immunosuppression for GVHD (e.g., corticosteroids, T-cell-depletion; tumor necrosis factor-□ inhibition) Inflammatory fungal pneumonia; angioinvasion with coagulative necrosis resembling aspergillosis classically associated with neutropenia may occur [2022]
Solid organ transplantation Immunosuppression to prevent allograft rejection May range from an acute inflammatory pneumonia to a chronic necrotizing aspergillosis; in lung transplant recipients, Aspergillus tracheobronchitis may affect the anastomotic site and cause dehiscence
Advanced AIDS CD4+ T-cell count generally < 100/ul; immunocompromising conditions (e.g., neutropenia) and other opportunistic infections often co-exist Acute to slowly progressive necrotizing pneumonia; variable histological findings: neutrophilic infiltrates, vascular invasion, walled-off abscesses and cavitation occur; extrapulmonary dissemination observed [23]
Chronic granulomatous disease Defective NADPH oxidase Varies from acute pneumonia to slowly progressive disease; pyogranulomatous inflammation without hyphal vascular invasion or coagulative necrosis; “mulch pneumonitis” is an acute hypersensitivity response to a large aerosolized exposure [24]
Pre-existing structural lung disease (e.g., emphysema, prior cavitary tuberculosis) Comorbid conditions, including diabetes, malnutrition, inhaled and low-dose systemic corticosteroids Chronic necrotizing pulmonary aspergillosis: slowly progressive invasive fungal pneumonia with inflammatory necrosis [25]
Aspergilloma Pre-existing structural lung diseases, e.g. bronchiectasis or prior cavitary tuberculosis “Fungal ball” composed of hyphal elements in pre-existing cavity; erosion into adjacent vessels can cause life-threatening hemoptysis; surgical resection is the definitive treatment for hemoptysis from aspergilloma
Allergic bronchopulmonary aspergillosis (ABPA) Allergic disease; can be an important complication of cystic fibrosis [26] Airway plugging with hyphae, mucous, and inflammatory cells; hyphae do not invade lung parenchyma; airway and lung hypereosinophilic inflammation; goblet cell hyperplasia; central bronchiecatasis in advanced disease

Table is adapted from [4].

Among allogeneic HSCT recipients, three periods of risk for invasive mould disease occur: 1) the neutropenic period following the conditioning regimen; 2) acute graft-versus-host disease (GVHD); and 3) chronic GVHD (after day 100 of transplant) [30]. Invasive mould disease is more common during intensive immunosuppressive therapy for GVHD than during neutropenia [3135]. This observation may be due to shortening of the neutropenic period following conditioning as a result of myelopoietic growth factors and use peripheral blood stem cell allografts that contain greater numbers of myeloid progenitors than bone marrow-derived allografts. T-cell depletion of allografts is also associated with a high risk of invasive aspergillosis and cytomegalovirus disease [36].

Among solid organ transplant recipients, the risk of invasive aspergillosis correlates with the intensity of immunosuppression used to prevent or treat allograft rejection. The incidence of invasive aspergillosis is highest after lung transplantation [37]. Colonization of the native lung with Aspergillus species, a common occurrence in end-stage lung disease, may be a source of fungal disease following single-lung transplantation [38,39]. Intensive immunosuppressive therapy used in non-transplant recipients, such as those with autoimmune diseases, can occasionally be complicated by aspergillosis [40].

There is also a growing appreciation of invasive aspergillosis in persons with less severe levels of immunocompromise. For example, chronic necrotizing pulmonary aspergillosis (CNPA) generally occurs in with patients pre-existing structural lung diseases such as prior tuberculosis or lung abscess or modest immune impairment, such as occurs with diabetes, poor nutrition, chronic obstructive pulmonary disease, or low-dose corticosteroids [25]. Fungal disease is slowly progressive over months to years, and may require life-long therapy. Mannose-binding lectin polymorphisms may be susceptibility factors for CNPA [41]. Invasive pulmonary aspergillosis has also been reported in critically ill patients without a documented systemic immune disease [4244].

Clinical Manifestations and Diagnosis

Invasive aspergillosis principally involves the sinopulmonary tract, a reflection of inhalation being the most common route of entry of Aspergillus spores (rarely, other sites of entry, such as the gastrointestinal tract or skin, occur). Fever, cough, and dyspnea are frequent, although non-specific, findings of pulmonary aspergillosis, the most common site of invasive aspergillosis. Vascular invasion may manifest as pleuritic chest pain due to pulmonary infarction or hemoptysis. Central nervous system involvement is a devastating consequence of disseminated aspergillosis, and may manifest with seizures or focal neurological signs from mass effect or stroke. Premature neonates can also develop aspergillosis, with the skin being the most common site of disease [45,46].

Diagnosis of invasive aspergillosis is difficult. Therefore in both clinical practice and research, different levels of certainty exist regarding the diagnosis of invasive aspergillosis. Demonstration of invasive hyphae histologically or a positive culture from a normally sterile environment (e.g., pleural fluid) is equated as proven invasive fungal disease. “Probable” invasive aspergillosis requires a combination of host factors that predispose to invasive aspergillosis (e.g., prolonged neutropenia, transplantation) and clinical (e.g., evidence of pneumonia) and mycological criteria [47]. These consensus criteria were designed for clinical research, but can be applied to clinical practice with the understanding that therapy for suspected invasive aspergillosis is commonly initiated prior to fulfilling these intentionally restrictive criteria.

In neutropenic patients, persistent fever may be the only sign of invasive fungal disease. A chest CT scan is more sensitive than radiographs for detection of early pulmonary aspergillosis [48], and should be considered in patients with 10 to 14 days of neutropenia (neutrophil count < 500/ul) and persistent or recurrent fever of unknown etiology unresponsive to empirical antibacterial agents [49]. The earliest radiological sign of invasive aspergillosis is a nodule[50]. A halo sign defined as a macronodule surrounded by a perimeter of ground-glass opacity corresponding to alveolar hemorrhage, is suggestive of invasive aspergillosis in patients with compatible host factors and initiation of treatment based on this sign has been associated with a better response compared to when therapy was initiated for more advanced fungal disease [51]. However other moulds and bacterial pathogens capable of angioinvasion (e.g., Pseudomonas aeruginosa) can produce a similar appearance. Other radiographic findings associated with invasive aspergillosis are consolidation, wedge-shaped infarcts, and cavitation. In contrast to adults, children with invasive pulmonary aspergillosis frequently do not manifest cavitation or the air crescent or halo signs [52].

Mycological criteria require either isolation of Aspergillus species from the sinopulmonary tract or positive antigen-based laboratory markers. Bronchoalveolar lavage fluid (BALF) cultures have at best 50% sensitivity in focal pulmonary lesions [53]. Antigen-based diagnosis relies on serum detection of either galactomannan or beta-D-glucan, two fungal cell wall constituents. The galactomannan assay is relatively specific for invasive aspergillosis, whereas the beta-D-glucan assay also detects other invasive fungal diseases, including candidiasis, other mould pathogens (excluding zygomycetes), and Pneumocystis jiroveci (formerly Pneumocystis carinii) [54,55].

The serum galactomannan assay has been principally studied in patients with leukemia and HSCT recipients, and its performance varies in different reports. In a meta-analysis, the serum galactomannan assay had a sensitivity of 71% and specificity of 89%, with significant heterogeneity in diagnostic accuracy among different patient groups [56]. Use of mould-active agents decreases the sensitivity of the galactomannan assay [57]. There are causes of false-positive results, including concomitant piperacillin/tazolbactam, other beta-lactam antibiotics, and gluconate-containing intravenous fluids. Cross-reactivity with other fungi (e.g., Histoplasma capsulatum) with similar cell wall galactomannan to Aspergillus can cause positive results [58]. Galactomannan detection in BALF appears to be more sensitive than serum detection [43,58], and can be used to support a diagnosis of probable aspergillosis [47]. As an alternative to bronchoscopy, percutaneous lung biopsy may be attempted for peripheral nodules. Thoracoscopic lung biopsy should be considered in a deteriorating patient when less invasive procedures produce negative results. Thrombocytopenia may limit the ability to perform invasive procedures.

In addition to serving as a diagnostic adjunct for invasive aspergillosis, a falling or rising serum galactomannan level may be useful as an early marker of therapeutic success or failure, respectively, in invasive aspergillosis [5961]. More research is required to validate the use of serum galactomannan monitoring as a therapeutic marker in different patient populations with invasive aspergillosis [62].

PCR-based diagnosis of invasive fungal diseases, although promising, is currently investigational. Potential advantages include rapidity, low cost, the ability to establish a diagnosis at the species level and to detect genes that confer antifungal resistance. Limitations include lack of standardized methods, difficulty in reliably distinguishing fungal colonization from disease, and the potential for contamination with fungal DNA [63].

Therapy for invasive aspergillosis

Voriconazole has become the gold standard as primary therapy for invasive aspergillosis [64] (Table 2).

Table 2.

Antifungal agents used to prevent and treat aspergillosis

Antifungal drug Dosing Comments
Voriconazole Adults: Intravenous (IV) 6 mg/kg Q12h × 2 doses, then 4 mg/kg Q12h; Oral 200 mg or rounded to 4 mg/kg twice daily
  • Superior to amphotercin B as primary therapy for invasive aspergillosis

  • Toxicities: reversible visual symptoms are common, but rarely require stopping drug; liver enzyme abnormalities, encephalopathy (uncommon)

  • Drug-drug interactions similar to other mould-active azoles

  • Significant inter-individual variability in systemic exposure; therapeutic drug monitoring can be considered

  • CYP 2C19 has genetic polymorphisms, with 15–20% of Asians expected to be slow metabolizers

  • IV formulation should be used with caution in patients with significant renal impairment (e.g., creatinine clearance < 50 ml/min) because of potential for systemic accumulation of the cyclodextrin vehicle that can, in turn, cause renal toxicity.

Pediatric: 7 mg/kg twice daily without a loading dose is approved by the European Medicines Agency (EMEA) in children aged 2 to 11 years; older children should be dosed as adults

Itraconazole - Oral capsules: 400 mg daily, which can be administered QD or divided b.i.d..
  • Oral solution has better bioavailability compared to the capsule

  • Therapeutic drug monitoring is advised, aiming for a trough of at least 250 ng/ml by HPLC

  • Contraindicated in patients with systolic cardiac dysfunction of a history of congestive heart failure

  • Effective in corticosteroid-dependent ABPA

  • IV formulation should be used with caution in patients with significant renal impairment because of potential for systemic accumulation of the cyclodextrin vehicle that can, in turn, cause renal toxicity.

-Oral solution: 2.5 mg/kg b.i.d.
- Pediatric oral dosing for children aged > 5 years: 2.5 mg/kg b.i.d; experience in younger children is lacking
- Intravenous (no longer marketed in U.S.) : 200 mg b.i.d. × 4 doses, then 200 mg daily

Posaconazole U.S. FDA-approved prophylactic dose: 200 mg T.I.D. in patients aged 13 years and over at high risk for invasive fungal disease
  • Currently, only available as an oral formulation

  • Variable serum levels

  • Must be taken with food or enteral nutrition; oral bioavailability is maximized when taken with fatty food

  • Effective as prophylaxis in patients with acute myelogenous leukemia and myelodysplastic syndrome receiving induction chemotherapy [65] and in patients with severe GVHD [66].


Amphotericin B deoxycholate Invasive mould diseases: 1.0 to 1.5 mg/kg daily
  • Significant infusional and nephrotoxicity can limit its use

  • Saline hydration may avert nephrotoxicity

  • Lipid formulations are generally better tolerated


Liposomal amphotericin B Adults: 3 mg/kg daily
  • 3 mg/kg/day as effective as but less toxic than 10 mg/kg/day as initial therapy for invasive aspergillosis in adults [68]

Clearance and volume of distribution influenced by body weight in pediatric oncology patients; higher mg/kg dosing may be optimal in patients weighing less than 20 kg [67]

Amphotericin B lipid complex 5 mg/kg daily
  • large non-randomized database for invasive aspergillosis [69]


Amphotericin B colloidal dispersion (ABCD) 5–6 mg/kg/day
  • ABCD (6 mg/kg/day) resulted in similar efficacy, lower nephrotoxicity, and greater infusional toxicity compared to Amb-D (1–1.5 mg/kg/day) as primary therapy for invasive aspergillosis [70]


Caspofungin - FDA-approved dose: 70 mg × 1, then 50 mg daily; dose of 70 mg daily can be considered for invasive aspergillosis [71]
  • Only echinocandin FDA-approved as salvage therapy for invasive aspergillosis

- In patients with moderate hepatic insufficiency (Child-Pugh score 7 to 9), a dose of 70 mg × 1, followed by 35 mg daily is advised by U.S. FDA.
- Pediatric dose: 50 mg/m2 (35 mg/m2 in patients with moderate liver disease) [72]

Micafungin - Prophylaxis in adult HSCT recipients during neutropenia: 50 mg daily; no approved dose as therapy for aspergillosis
  • Non-randomized trial showed safety of micafungin alone and in combination in patients with invasive aspergillosis [73]


Anidulafungin - no approved dose as therapy for aspergillosis
  • No clinical trial data as therapy for invasive aspergillosis

  • randomized trial underway comparing voriconazole + anidulafungin with voriconazole alone as primary therapy for invasive aspergillosis

Voriconazole was more effective than amphotericin B deoxycholate (AmB-D) as initial therapy for invasive aspergillosis and was associated with significantly improved survival (71% vs. 58%, respectively) in a randomized trial [51]. The rate of successful outcomes was superior in voriconazole compared to AmB-D recipients (53% versus 32% respectively). The poorest prognosis occurred in extrapulmonary aspergillosis and in allogeneic HSCT recipients.

Voriconazole exhibits non-linear elimination in adults. It is estimated that increasing the oral dose of voriconazole from 200 mg Q12h to 300 mg Q12h leads to a 2.5-fold increase in exposure, and increasing the intravenous dose from 3 mg/kg Q12h to 4 mg/kg Q12h results in a 2.3-fold increase in exposure (package insert). In contrast, clearance of voriconazole in children is linear, necessitating higher dosing per kg of body weight in children to achieve comparable exposure as adults [74,75].

There can be considerable inter-individual variability and intra-individual variability over time in voriconazole exposure [76,77]. Small studies have noted a relationship between low plasma voriconazole levels and failure of therapy of invasive mycoses [78,79], and between high voriconazole levels and toxicity [79,80]. Therapeutic drug monitoring for voriconazole should be considered, particularly in cases of refractory fungal disease or drug toxicity. Therapeutic drug monitoring is however limited by lack of well-defined reference ranges for serum drug levels. In cases of suspected therapeutic failure, aiming for a serum voriconazole of at least 2 ug/ml is reasonable based on limited published data is reasonable [78].

Mould-active azoles and ketoconazole are potent inhibitors of cytochrome P450 3A4 (CYP3A4), which catalyzes the initial step in the clearance of several drugs. Inhibition of the CYP3A4 isoenzyme by azoles accounts for the majority of drug-drug interactions. Certain drugs metabolized by CYP3A4 require dose reduction when co-administered with voriconazole (e.g., calcineurin inhibitors).

Resistance of Aspergillus species to antifungal agents is a possible cause of failure of therapy. Aspergillus terreus is resistant to amphotericin B [81]. Mutations in the cyp51A gene of A. fumigatus have been linked to resistance to itraconazole; isolates harboring this mutation as also less sensitive to other mould-active azoles [82].

For patients with invasive aspergillosis refractory to voriconazole or who are intolerant of voriconazole, a lipid formulation of amphotericin B or an echinocandin can be used. Lipid formulations are generally preferred over amphotericin B deoxycholate for invasive aspergillosis, given that prolonged use of the conventional formulation is poorly tolerated because of nephrotoxicity. Of the echinocandins, caspofungin is approved by the U.S. FDA as salvage therapy for invasive aspergillosis. Caspofungin monotherapy led to a successful outcome in 37 (45%) of 83 patients with invasive aspergillosis, 86% of whom had disease refractory to standard antifungal therapy [83].

There is substantial interest in pairing cell wall-active echinocandins with either amphotericin B formulations or mould-active azoles as therapy for invasive aspergillosis. In one provocative study, Marr et al.[84] reported a survival advantage of voriconazole plus caspofungin compared to voriconazole alone in a retrospective analysis of salvage therapy for invasive aspergillosis. However, this database involved small numbers of patients, and the two groups were non-contemporaneous; therefore, other host and infection-related factors may have influenced the outcome. A randomized trial comparing voriconazole (the gold standard) with voriconazole and anidulafungin (an echinocandin) has been initiated.

When feasible, immunosuppressive therapy (e.g., corticosteroids) should be reduced or discontinued. Adjunctive myeloid colony-stimulating factors (G-CSF or GM-CSF) should be considered in neutropenic patients with severe infections, such as aspergillosis [85]. Granulocyte transfusions in neutropenic patients can be considered in refractory or disseminated aspergillosis, realizing that their benefit versus toxicity risk has not been established [86]. Although marrow recovery is critically important for host defense against aspergillosis, myeloid recovery may lead to the liquefaction of pulmonary foci and increase the risk of pulmonary hemorrhage [87] and may increase pulmonary consolidation as part of an immune reconstitution syndrome in patients with invasive aspergillosis [88,89]. Adjunctive recombinant interferon-γ, which activates neutrophils and monocytes, can also be considered in cases of severe or refractory aspergillosis [90].

Future perspectives

We are gaining more knowledge about how the host response to Aspergillus species that may prove to be useful in developing novel prevention and treatment strategies. In addition to the severe forms of immune impairment that predispose to invasive aspergillosis (e.g., prolonged neutropenia, immunosuppressive regiments used in transplantation), variations in specific host defense alleles also likely affect risk offungal disease. As examples, Bochud et al. [91] showed that donor TLR4 polymorphisms affected the risk of invasive aspergillosis in allogeneic HSCT recipients. Zaas et al. [92] showed in both mouse models of aspergillosis and in human allogeneic HSCT recipients that polymorphisms in the plasminogen allele affected the risk of aspergillosis. Such studies may be useful to more precisely stratify immunocompromised patients regarding risk of aspergillosis, and potentially use more targeted approaches to antifungal prophylaxis.

We are also learning more about the biology of Aspergillus species that might be exploited therapeutically. Fungal genomic studies have shown that members of the Aspergillus genus are extremely diverse. The level of molecular divergence among certain Aspergillus species is similar to the divergence between humans and fish [93]. Although most cases of invasive aspergillosis are caused by A. fumigatus, it represents a small minority of aerosolized spores that would be in contact with human respiratory mucosa. Therefore, one of the major benefits of comparative genomics among pathogenic and non-pathogenic Aspergillus species will be to identify pathways (virulence factors) that enable disease to occur in susceptible hosts, and to identify novel targets for drug development. The recent discovery of a sexual cycle in A. fumigatus provides an invaluable tool top study the genetic basis of pathogenicity and antifungal resistance in A. fumigatus [94].

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • 1.Cornell MJ, Alam I, Soanes DM, Wong HM, Hedeler C, Paton NW, Rattray M, Hubbard SJ, Talbot NJ, Oliver SG. Comparative genome analysis across a kingdom of eukaryotic organisms: specialization and diversification in the fungi. Genome Res. 2007;17:1809–1822. doi: 10.1101/gr.6531807. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Casadevall A, Steenbergen JN, Nosanchuk JD. `Ready made' virulence and `dual use' virulence factors in pathogenic environmental fungi--the Cryptococcus neoformans paradigm. Curr Opin Microbiol. 2003;6:332–337. doi: 10.1016/s1369-5274(03)00082-1. [DOI] [PubMed] [Google Scholar]
  • 3.Lemaitre B, Nicolas E, Michaut L, Reichhart JM, Hoffmann JA. The dorsoventral regulatory gene cassette spatzle/Toll/cactus controls the potent antifungal response in Drosophila adults. Cell. 1996;86:973–983. doi: 10.1016/s0092-8674(00)80172-5. [DOI] [PubMed] [Google Scholar]
  • *4.Segal BH. Aspergillosis. N Engl J Med. 2009;360:1870–1884. doi: 10.1056/NEJMra0808853. [DOI] [PubMed] [Google Scholar]
  • 5.Schaffner A, Douglas H, Braude A. Selective protection against conidia by mononuclear and against mycelia by polymorphonuclear phagocytes in resistance to Aspergillus. Observations on these two lines of defense in vivo and in vitro with human and mouse phagocytes. J Clin Invest. 1982;69:617–631. doi: 10.1172/JCI110489. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Hohl TM, Van Epps HL, Rivera A, Morgan LA, Chen PL, Feldmesser M, Pamer EG. Aspergillus fumigatus triggers inflammatory responses by stage-specific beta-glucan display. PLoS Pathog. 2005;1:e30. doi: 10.1371/journal.ppat.0010030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Gersuk GM, Underhill DM, Zhu L, Marr KA. Dectin-1 and TLRs permit macrophages to distinguish between different Aspergillus fumigatus cellular states. J Immunol. 2006;176:3717–3724. doi: 10.4049/jimmunol.176.6.3717. [DOI] [PubMed] [Google Scholar]
  • 8.Steele C, Rapaka RR, Metz A, Pop SM, Williams DL, Gordon S, Kolls JK, Brown GD. The Beta-Glucan Receptor Dectin-1 Recognizes Specific Morphologies of Aspergillus fumigatus. PLoS Pathog. 2005;1:e42. doi: 10.1371/journal.ppat.0010042. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • **9.Aimanianda V, Bayry J, Bozza S, Kniemeyer O, Perruccio K, Elluru SR, Clavaud C, Paris S, Brakhage AA, Kaveri SV, Romani L, Latgé JP. Surface hydrophobin prevents immune recognition of airborne fungal spores. Nature. 2009;460(7259):1117–21. doi: 10.1038/nature08264. [DOI] [PubMed] [Google Scholar]; This study identified a hydrophobic protein expressed only at the spore stage of A. fumigatus that silences the immune response.
  • 10.Gantner BN, Simmons RM, Canavera SJ, Akira S, Underhill DM. Collaborative induction of inflammatory responses by dectin-1 and Toll-like receptor 2. J Exp Med. 2003;197:1107–1117. doi: 10.1084/jem.20021787. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Segal BH, DeCarlo ES, Kwon-Chung KJ, Malech HL, Gallin JI, Holland SM. Aspergillus nidulans infection in chronic granulomatous disease. Medicine (Baltimore) 1998;77:345–354. doi: 10.1097/00005792-199809000-00004. [DOI] [PubMed] [Google Scholar]
  • *12.Reeves EP, Lu H, Jacobs HL, Messina CG, Bolsover S, Gabella G, Potma EO, Warley A, Roes J, Segal AW. Killing activity of neutrophils is mediated through activation of proteases by K+ flux. Nature. 2002;416:291–297. doi: 10.1038/416291a. [DOI] [PubMed] [Google Scholar]
  • *13.Holland SM, Vinh DC. Yeast infections--human genetics on the rise. N Engl J Med. 2009;361:1798–1801. doi: 10.1056/NEJMe0907186. [DOI] [PubMed] [Google Scholar]
  • *14.Gaffen SL. Structure and signalling in the IL-17 receptor family. Nat Rev Immunol. 2009;9:556–567. doi: 10.1038/nri2586. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • *15.Zelante T, De Luca A, D'Angelo C, Moretti S, Romani L. IL-17/Th17 in anti-fungal immunity: what's new? Eur J Immunol. 2009;39:645–648. doi: 10.1002/eji.200839102. [DOI] [PubMed] [Google Scholar]
  • **16.Ferwerda B, Ferwerda G, Plantinga TS, Willment JA, van Spriel AB, Venselaar H, Elbers CC, Johnson MD, Cambi A, Huysamen C, et al. Human dectin-1 deficiency and mucocutaneous fungal infections. N Engl J Med. 2009;361:1760–1767. doi: 10.1056/NEJMoa0901053. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • **17.Glocker EO, Hennigs A, Nabavi M, Schaffer AA, Woellner C, Salzer U, Pfeifer D, Veelken H, Warnatz K, Tahami F, et al. A homozygous CARD9 mutation in a family with susceptibility to fungal infections. N Engl J Med. 2009;361:1727–1735. doi: 10.1056/NEJMoa0810719. [DOI] [PMC free article] [PubMed] [Google Scholar]; References 17 and 18 show that familial mucocutaneous candidiasis is linked to deficiencies in signaling of Dectin-1, a pathogen recognition factor that recognizes fungal beta-glucans.
  • **18.Werner JL, Metz AE, Horn D, Schoeb TR, Hewitt MM, Schwiebert LM, Faro-Trindade I, Brown GD, Steele C. Requisite role for the dectin-1 beta-glucan receptor in pulmonary defense against Aspergillus fumigatus. J Immunol. 2009;182:4938–4946. doi: 10.4049/jimmunol.0804250. [DOI] [PMC free article] [PubMed] [Google Scholar]; This study showed a protective role of Dectin-1 in host defense and inflammation in aspergillosis in mice.
  • **19.Romani L, Fallarino F, De Luca A, Montagnoli C, D'Angelo C, Zelante T, Vacca C, Bistoni F, Fioretti MC, Grohmann U, et al. Defective tryptophan catabolism underlies inflammation in mouse chronic granulomatous disease. Nature. 2008;451:211–215. doi: 10.1038/nature06471. [DOI] [PubMed] [Google Scholar]; This study showed that NADPH oxidase mediates host defense and inflammation in aspergillosis in mice through activation of tryptophan catabolism.
  • 20.Chamilos G, Luna M, Lewis RE, Bodey GP, Chemaly R, Tarrand JJ, Safdar A, Raad II, Kontoyiannis DP. Invasive fungal infections in patients with hematologic malignancies in a tertiary care cancer center: an autopsy study over a 15-year period (1989–2003) Haematologica. 2006;91:986–989. [PubMed] [Google Scholar]
  • 21.Shaukat A, Bakri F, Young P, Hahn T, Ball D, Baer MR, Wetzler M, Slack JL, Loud P, Czuczman M, et al. Invasive filamentous fungal infections in allogeneic hematopoietic stem cell transplant recipients after recovery from neutropenia: Clinical, radiologic, and pathologic characteristics. Mycopathologia. 2005;159:181–188. doi: 10.1007/s11046-004-5495-0. [DOI] [PubMed] [Google Scholar]
  • 22.Stergiopoulou T, Meletiadis J, Roilides E, Kleiner DE, Schaufele R, Roden M, Harrington S, Dad L, Segal B, Walsh TJ. Host-dependent patterns of tissue injury in invasive pulmonary aspergillosis. Am J Clin Pathol. 2007;127:349–355. doi: 10.1309/UJRV9DLC11RM3G8R. [DOI] [PubMed] [Google Scholar]
  • 23.Nash G, Irvine R, Kerschmann RL, Herndier B. Pulmonary aspergillosis in acquired immune deficiency syndrome: autopsy study of an emerging pulmonary complication of human immunodeficiency virus infection. Hum Pathol. 1997;28:1268–1275. doi: 10.1016/s0046-8177(97)90200-8. [DOI] [PubMed] [Google Scholar]
  • 24.Siddiqui S, Anderson VL, Hilligoss DM, Abinun M, Kuijpers TW, Masur H, Witebsky FG, Shea YR, Gallin JI, Malech HL, et al. Fulminant mulch pneumonitis: an emergency presentation of chronic granulomatous disease. Clin Infect Dis. 2007;45:673–681. doi: 10.1086/520985. [DOI] [PubMed] [Google Scholar]
  • 25.Binder RE, Faling LJ, Pugatch RD, Mahasaen C, Snider GL. Chronic necrotizing pulmonary aspergillosis: a discrete clinical entity. Medicine (Baltimore) 1982;61:109–124. doi: 10.1097/00005792-198203000-00005. [DOI] [PubMed] [Google Scholar]
  • 26.Stevens DA, Moss RB, Kurup VP, Knutsen AP, Greenberger P, Judson MA, Denning DW, Crameri R, Brody AS, Light M, et al. Allergic bronchopulmonary aspergillosis in cystic fibrosis--state of the art: Cystic Fibrosis Foundation Consensus Conference. Clin Infect Dis. 2003;37(Suppl 3):S225–264. doi: 10.1086/376525. [DOI] [PubMed] [Google Scholar]
  • 27.Segal BH, Walsh TJ. Current approaches to diagnosis and treatment of invasive aspergillosis. Am J Respir Crit Care Med. 2006;173:707–717. doi: 10.1164/rccm.200505-727SO. [DOI] [PubMed] [Google Scholar]
  • 28.Introduction- The Aspergillus fumigatus genome database on World Wide Web URL:
  • 29.McNeil MM, Nash SL, Hajjeh RA, Phelan MA, Conn LA, Plikaytis BD, Warnock DW. Trends in mortality due to invasive mycotic diseases in the United States, 1980–1997. Clin Infect Dis. 2001;33:641–647. doi: 10.1086/322606. Epub 2001 Jul 2030. [DOI] [PubMed] [Google Scholar]
  • 30.Marty FM, Rubin RH. The prevention of infection post-transplant: the role of prophylaxis, preemptive and empiric therapy. Transpl Int. 2006;19:2–11. doi: 10.1111/j.1432-2277.2005.00218.x. [DOI] [PubMed] [Google Scholar]
  • 31.Bhatti Z, Shaukat A, Almyroudis NG, Segal BH. Review of epidemiology, diagnosis, and treatment of invasive mould infections in allogeneic hematopoietic stem cell transplant recipients. Mycopathologia. 2006;162:1–15. doi: 10.1007/s11046-006-0025-x. [DOI] [PubMed] [Google Scholar]
  • 32.Yuen KY, Woo PC, Ip MS, Liang RH, Chiu EK, Siau H, Ho PL, Chen FF, Chan TK. Stage-specific manifestation of mold infections in bone marrow transplant recipients: risk factors and clinical significance of positive concentrated smears. Clin Infect Dis. 1997;25:37–42. doi: 10.1086/514492. [DOI] [PubMed] [Google Scholar]
  • 33.Marr KA, Carter RA, Boeckh M, Martin P, Corey L. Invasive aspergillosis in allogeneic stem cell transplant recipients: changes in epidemiology and risk factors. Blood. 2002;100:4358–4366. doi: 10.1182/blood-2002-05-1496. [DOI] [PubMed] [Google Scholar]
  • 34.Jantunen E, Ruutu P, Niskanen L, Volin L, Parkkali T, Koukila-Kahkola P, Ruutu T. Incidence and risk factors for invasive fungal infections in allogeneic BMT recipients. Bone Marrow Transplant. 1997;19:801–808. doi: 10.1038/sj.bmt.1700737. [DOI] [PubMed] [Google Scholar]
  • 35.Wald A, Leisenring W, van Burik JA, Bowden RA. Epidemiology of Aspergillus infections in a large cohort of patients undergoing bone marrow transplantation. J Infect Dis. 1997;175:1459–1466. doi: 10.1086/516480. [DOI] [PubMed] [Google Scholar]
  • 36.van Burik JA, Carter SL, Freifeld AG, High KP, Godder KT, Papanicolaou GA, Mendizabal AM, Wagner JE, Yanovich S, Kernan NA. Higher risk of cytomegalovirus and aspergillus infections in recipients of T cell-depleted unrelated bone marrow: analysis of infectious complications in patients treated with T cell depletion versus immunosuppressive therapy to prevent graft-versus-host disease. Biol Blood Marrow Transplant. 2007;13:1487–1498. doi: 10.1016/j.bbmt.2007.08.049. [DOI] [PubMed] [Google Scholar]
  • 37.Morgan J, Wannemuehler KA, Marr KA, Hadley S, Kontoyiannis DP, Walsh TJ, Fridkin SK, Pappas PG, Warnock DW. Incidence of invasive aspergillosis following hematopoietic stem cell and solid organ transplantation: interim results of a prospective multicenter surveillance program. Med Mycol. 2005;43(Suppl 1):S49–58. doi: 10.1080/13693780400020113. [DOI] [PubMed] [Google Scholar]
  • 38.Hadjiliadis D, Sporn TA, Perfect JR, Tapson VF, Davis RD, Palmer SM. Outcome of lung transplantation in patients with mycetomas. Chest. 2002;121:128–134. doi: 10.1378/chest.121.1.128. [DOI] [PubMed] [Google Scholar]
  • 39.Westney GE, Kesten S, De Hoyos A, Chapparro C, Winton T, Maurer JR. Aspergillus infection in single and double lung transplant recipients. Transplantation. 1996;61:915–919. doi: 10.1097/00007890-199603270-00013. [DOI] [PubMed] [Google Scholar]
  • 40.Segal BH, Sneller MC. Infectious complications of immunosuppressive therapy in patients with rheumatic diseases. Rheum Dis Clin North Am. 1997;23:219–237. doi: 10.1016/s0889-857x(05)70327-6. [DOI] [PubMed] [Google Scholar]
  • 41.Crosdale DJ, Poulton KV, Ollier WE, Thomson W, Denning DW. Mannose-binding lectin gene polymorphisms as a susceptibility factor for chronic necrotizing pulmonary aspergillosis. J Infect Dis. 2001;184:653–656. doi: 10.1086/322791. [DOI] [PubMed] [Google Scholar]
  • 42.Meersseman W, Lagrou K, Maertens J, Van Wijngaerden E. Invasive aspergillosis in the intensive care unit. Clin Infect Dis. 2007;45:205–216. doi: 10.1086/518852. [DOI] [PubMed] [Google Scholar]
  • 43.Meersseman W, Lagrou K, Maertens J, Wilmer A, Hermans G, Vanderschueren S, Spriet I, Verbeken E, Van Wijngaerden E. Galactomannan in bronchoalveolar lavage fluid: a tool for diagnosing aspergillosis in intensive care unit patients. Am J Respir Crit Care Med. 2008;177:27–34. doi: 10.1164/rccm.200704-606OC. [DOI] [PubMed] [Google Scholar]; This study showed the utility of bronchoalveolar lavage fluid galactomannan testing in diagnosing invasive aspergillosis in critically ill patients without classical risk factors for aspergillosis
  • 44.Vandewoude KH, Blot SI, Depuydt P, Benoit D, Temmerman W, Colardyn F, Vogelaers D. Clinical relevance of Aspergillus isolation from respiratory tract samples in critically ill patients. Crit Care. 2006;10:R31. doi: 10.1186/cc4823. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Horii KA, Nopper AJ. Emerging cutaneous infections in the premature neonate. Adv Dermatol. 2007;23:177–195. doi: 10.1016/j.yadr.2007.07.012. [DOI] [PubMed] [Google Scholar]
  • 46.Singer S, Singer D, Ruchel R, Mergeryan H, Schmidt U, Harms K. Outbreak of systemic aspergillosis in a neonatal intensive care unit. Mycoses. 1998;41:223–227. doi: 10.1111/j.1439-0507.1998.tb00328.x. [DOI] [PubMed] [Google Scholar]
  • 47.De Pauw B, Walsh TJ, Donnelly JP, Stevens DA, Edwards JE, Calandra T, Pappas PG, Maertens J, Lortholary O, Kauffman CA, 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;46:1813–1821. doi: 10.1086/588660. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Caillot D, Casasnovas O, Bernard A, Couaillier JF, Durand C, Cuisenier B, Solary E, Piard F, Petrella T, Bonnin A, et al. Improved management of invasive pulmonary aspergillosis in neutropenic patients using early thoracic computed tomographic scan and surgery. J Clin Oncol. 1997;15:139–147. doi: 10.1200/JCO.1997.15.1.139. [DOI] [PubMed] [Google Scholar]
  • 49.Segal BH, Freifeld AG, Baden LR, Brown AE, Casper C, Dubberke E, Gelfand M, Greene JN, Ison MG, Ito JI, et al. Prevention and treatment of cancer-related infections. J Natl Compr Canc Netw. 2008;6:122–174. doi: 10.6004/jnccn.2008.0013. [DOI] [PubMed] [Google Scholar]; National Comprehensive Cancer network (NCCN) guidelines on prevention, diagnosis, and treatment of infectious complciations in patients with cancer. Yearly updates are posted online (www.nccn.org)
  • 50.Greene RE, Schlamm HT, Oestmann JW, Stark P, Durand C, Lortholary O, Wingard JR, Herbrecht R, Ribaud P, Patterson TF, et al. Imaging findings in acute invasive pulmonary aspergillosis: clinical significance of the halo sign. Clin Infect Dis. 2007;44:373–379. doi: 10.1086/509917. [DOI] [PubMed] [Google Scholar]
  • *51.Herbrecht R, Denning DW, Patterson TF, Bennett JE, Greene RE, Oestmann JW, Kern WV, Marr KA, Ribaud P, Lortholary O, et al. Voriconazole versus amphotericin B for primary therapy of invasive aspergillosis. N Engl J Med. 2002;347:408–415. doi: 10.1056/NEJMoa020191. [DOI] [PubMed] [Google Scholar]
  • **52.Burgos A, Zaoutis TE, Dvorak CC, Hoffman JA, Knapp KM, Nania JJ, Prasad P, Steinbach WJ. Pediatric invasive aspergillosis: a multicenter retrospective analysis of 139 contemporary cases. Pediatrics. 2008;121:e1286–1294. doi: 10.1542/peds.2007-2117. [DOI] [PubMed] [Google Scholar]; This multicenter retrospective study shows diagnostically relevant differences in features of aspergillosis in children versus adults.
  • 53.Levine SJ. An approach to the diagnosis of pulmonary infections in immunosuppressed patients. Semin Respir Infect. 1992;7:81–95. [PubMed] [Google Scholar]
  • 54.Odabasi Z, Mattiuzzi G, Estey E, Kantarjian H, Saeki F, Ridge RJ, Ketchum PA, Finkelman MA, Rex JH, Ostrosky-Zeichner L. Beta-D-glucan as a diagnostic adjunct for invasive fungal infections: validation, cutoff development, and performance in patients with acute myelogenous leukemia and myelodysplastic syndrome. Clin Infect Dis. 2004;39:199–205. doi: 10.1086/421944. Epub 2004 Jun 2028. [DOI] [PubMed] [Google Scholar]
  • 55.Marty FM, Koo S, Bryar J, Baden LR. (1–>3)beta-D-glucan assay positivity in patients with Pneumocystis (carinii) jiroveci pneumonia. Ann Intern Med. 2007;147:70–72. doi: 10.7326/0003-4819-147-1-200707030-00018. [DOI] [PubMed] [Google Scholar]
  • 56.Pfeiffer CD, Fine JP, Safdar N. Diagnosis of invasive aspergillosis using a galactomannan assay: a meta-analysis. Clin Infect Dis. 2006;42:1417–1727. doi: 10.1086/503427. [DOI] [PubMed] [Google Scholar]
  • 57.Marr KA, Balajee SA, McLaughlin L, Tabouret M, Bentsen C, Walsh TJ. Detection of galactomannan antigenemia by enzyme immunoassay for the diagnosis of invasive aspergillosis: variables that affect performance. J Infect Dis. 2004;190:641–649. doi: 10.1086/422009. Epub 2004 Jul 2001. [DOI] [PubMed] [Google Scholar]
  • 58.Wheat LJ, Walsh TJ. Diagnosis of invasive aspergillosis by galactomannan antigenemia detection using an enzyme immunoassay. Eur J Clin Microbiol Infect Dis. 2008;27:245–251. doi: 10.1007/s10096-007-0437-7. [DOI] [PubMed] [Google Scholar]
  • **59.Miceli MH, Grazziutti ML, Woods G, Zhao W, Kocoglu MH, Barlogie B, Anaissie E. Strong correlation between serum aspergillus galactomannan index and outcome of aspergillosis in patients with hematological cancer: clinical and research implications. Clin Infect Dis. 2008;46:1412–1422. doi: 10.1086/528714. [DOI] [PubMed] [Google Scholar]
  • **60.Woods G, Miceli MH, Grazziutti ML, Zhao W, Barlogie B, Anaissie E. Serum Aspergillus galactomannan antigen values strongly correlate with outcome of invasive aspergillosis: a study of 56 patients with hematologic cancer. Cancer. 2007;110:830–834. doi: 10.1002/cncr.22863. [DOI] [PubMed] [Google Scholar]
  • ** 61.Maertens J, Buve K, Theunissen K, Meersseman W, Verbeken E, Verhoef G, Van Eldere J, Lagrou K. Galactomannan serves as a surrogate endpoint for outcome of pulmonary invasive aspergillosis in neutropenic hematology patients. Cancer. 2009;115:355–362. doi: 10.1002/cncr.24022. [DOI] [PubMed] [Google Scholar]; References 5961 show the potential benefit of serial serum galactomannan monitoring as an early marker of response to antifungal treatment in invasive aspergillosis.
  • *62.Segal BH, Herbrecht R, Stevens DA, Ostrosky-Zeichner L, Sobel J, Viscoli C, Walsh TJ, Maertens J, Patterson TF, Perfect JR, et al. Defining responses to therapy and study outcomes in clinical trials of invasive fungal diseases: Mycoses Study Group and European Organization for Research and Treatment of Cancer consensus criteria. Clin Infect Dis. 2008;47:674–683. doi: 10.1086/590566. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • *63.Einsele H, Loeffler J. Contribution of new diagnostic approaches to antifungal treatment plans in high-risk haematology patients. Clin Microbiol Infect. 2008;14(Suppl 4):37–45. doi: 10.1111/j.1469-0691.2008.01980.x. [DOI] [PubMed] [Google Scholar]
  • **64.Walsh TJ, Anaissie EJ, Denning DW, Herbrecht R, Kontoyiannis DP, Marr KA, Morrison VA, Segal BH, Steinbach WJ, Stevens DA, et al. Treatment of aspergillosis: clinical practice guidelines of the Infectious Diseases Society of America. Clin Infect Dis. 2008;46:327–360. doi: 10.1086/525258. [DOI] [PubMed] [Google Scholar]; Authoritative guidelines from the Infectious Diseases Society of America (IDSA) on diagnosis and treatment of aspergillosis.
  • *65.Cornely OA, Maertens J, Winston DJ, Perfect J, Ullmann AJ, Walsh TJ, Helfgott D, Holowiecki J, Stockelberg D, Goh YT, et al. Posaconazole vs. fluconazole or itraconazole prophylaxis in patients with neutropenia. N Engl J Med. 2007;356:348–359. doi: 10.1056/NEJMoa061094. [DOI] [PubMed] [Google Scholar]
  • *66.Ullmann AJ, Lipton JH, Vesole DH, Chandrasekar P, Langston A, Tarantolo SR, Greinix H, Morais de Azevedo W, Reddy V, Boparai N, et al. Posaconazole or fluconazole for prophylaxis in severe graft-versus-host disease. N Engl J Med. 2007;356:335–347. doi: 10.1056/NEJMoa061098. [DOI] [PubMed] [Google Scholar]
  • 67.Hong Y, Shaw PJ, Nath CE, Yadav SP, Stephen KR, Earl JW, McLachlan AJ. Population pharmacokinetics of liposomal amphotericin B in pediatric patients with malignant diseases. Antimicrob Agents Chemother. 2006;50:935–942. doi: 10.1128/AAC.50.3.935-942.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Cornely OA, Maertens J, Bresnik M, Ebrahimi R, Ullmann AJ, Bouza E, Heussel CP, Lortholary O, Rieger C, Boehme A, 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;44:1289–1297. doi: 10.1086/514341. [DOI] [PubMed] [Google Scholar]
  • 69.Chandrasekar PH, Ito JI. Amphotericin B lipid complex in the management of invasive aspergillosis in immunocompromised patients. Clin Infect Dis. 2005;40(Suppl 6):S392–400. doi: 10.1086/429333. [DOI] [PubMed] [Google Scholar]
  • 70.Bowden R, Chandrasekar P, White MH, Li X, Pietrelli L, Gurwith M, van Burik JA, Laverdiere M, Safrin S, Wingard JR. A double-blind, randomized, controlled trial of amphotericin B colloidal dispersion versus amphotericin B for treatment of invasive aspergillosis in immunocompromised patients. Clin Infect Dis. 2002;35:359–366. doi: 10.1086/341401. Epub 2002 Jul 2025. [DOI] [PubMed] [Google Scholar]
  • 71.Maertens J, Glasmacher A, Herbrecht R, Thiebaut A, Cordonnier C, Segal BH, Killar J, Taylor A, Kartsonis N, Patterson TF, et al. Multicenter, noncomparative study of caspofungin in combination with other antifungals as salvage therapy in adults with invasive aspergillosis. Cancer. 2006;107:2888–2897. doi: 10.1002/cncr.22348. [DOI] [PubMed] [Google Scholar]
  • 72.Walsh TJ, Adamson PC, Seibel NL, Flynn PM, Neely MN, Schwartz C, Shad A, Kaplan SL, Roden MM, Stone JA, et al. Pharmacokinetics, safety, and tolerability of caspofungin in children and adolescents. Antimicrob Agents Chemother. 2005;49:4536–4545. doi: 10.1128/AAC.49.11.4536-4545.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Denning DW, Marr KA, Lau WM, Facklam DP, Ratanatharathorn V, Becker C, Ullmann AJ, Seibel NL, Flynn PM, van Burik JA, et al. Micafungin (FK463), alone or in combination with other systemic antifungal agents, for the treatment of acute invasive aspergillosis. J Infect. 2006;53:337–349. doi: 10.1016/j.jinf.2006.03.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Walsh TJ, Driscoll TA, Groll AH, Arrieta AC, Klein N, Bradley J, Jafri HS, Schlamm HT, Troke PF, Wood N, et al. Population Pharmacokinetic (POP-PK) analysis of voriconazole (VRC): developing a rationale for dosage in pediatric patients. 47th Interscience Conference on Antimicrobial Agents and Chemotherapy; Chicago, IL (M-620). Sept., 2007.2007. [Google Scholar]
  • 75.Walsh TJ, Karlsson MO, Driscoll T, Arguedas AG, Adamson P, Saez-Llorens X, Vora AJ, Arrieta AC, Blumer J, Lutsar I, et al. Pharmacokinetics and safety of intravenous voriconazole in children after single- or multiple-dose administration. Antimicrob Agents Chemother. 2004;48:2166–2172. doi: 10.1128/AAC.48.6.2166-2172.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • *76.Pascual A, Nieth V, Calandra T, Bille J, Bolay S, Decosterd LA, Buclin T, Majcherczyk PA, Sanglard D, Marchetti O. Variability of voriconazole plasma levels measured by new high-performance liquid chromatography and bioassay methods. Antimicrob Agents Chemother. 2007;51:137–143. doi: 10.1128/AAC.00957-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • *77.Trifilio S, Pennick G, Pi J, Zook J, Golf M, Kaniecki K, Singhal S, Williams S, Winter J, Tallman M, et al. Monitoring plasma voriconazole levels may be necessary to avoid subtherapeutic levels in hematopoietic stem cell transplant recipients. Cancer. 2007;109:1532–1535. doi: 10.1002/cncr.22568. [DOI] [PubMed] [Google Scholar]
  • *78.Smith J, Safdar N, Knasinski V, Simmons W, Bhavnani SM, Ambrose PG, Andes D. Voriconazole therapeutic drug monitoring. Antimicrob Agents Chemother. 2006;50:1570–1572. doi: 10.1128/AAC.50.4.1570-1572.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • **79.Pascual A, Calandra T, Bolay S, Buclin T, Bille J, Marchetti O. Voriconazole therapeutic drug monitoring in patients with invasive mycoses improves efficacy and safety outcomes. Clin Infect Dis. 2008;46:201–211. doi: 10.1086/524669. [DOI] [PubMed] [Google Scholar]; This paper and others showed substantial inter-individual variability in serum voriconazole levels, and showed a correlation between low levels an dtherapeutic failure and between high levels and drug toxicity.
  • *80.Tan K, Brayshaw N, Tomaszewski K, Troke P, Wood N. Investigation of the potential relationships between plasma voriconazole concentrations and visual adverse events or liver function test abnormalities. J Clin Pharmacol. 2006;46:235–243. doi: 10.1177/0091270005283837. [DOI] [PubMed] [Google Scholar]
  • 81.Steinbach WJ, Benjamin DK, Jr., Kontoyiannis DP, Perfect JR, Lutsar I, Marr KA, Lionakis MS, Torres HA, Jafri H, Walsh TJ. Infections due to Aspergillus terreus: a multicenter retrospective analysis of 83 cases. Clin Infect Dis. 2004;39:192–198. doi: 10.1086/421950. Epub 2004 Jun 2028. [DOI] [PubMed] [Google Scholar]
  • **82.Snelders E, van der Lee HA, Kuijpers J, Rijs AJ, Varga J, Samson RA, Mellado E, Donders AR, Melchers WJ, Verweij PE. Emergence of azole resistance in Aspergillus fumigatus and spread of a single resistance mechanism. PLoS Med. 2008;5:e219. doi: 10.1371/journal.pmed.0050219. [DOI] [PMC free article] [PubMed] [Google Scholar]; This study showed an increased frequency of itraconazole-resistant clincal Aspergillus fumigatus isolates over time. A substitution of leucine 98 for histidine in the cyp51A gene, together with two copies of a 34-bp sequence in tandem in the gene promoter (TR/L98H), was the principal resistance mechanism.
  • 83.Maertens J, Raad I, Petrikkos G, Boogaerts M, Selleslag D, Petersen FB, Sable CA, Kartsonis NA, Ngai A, Taylor A, et al. Efficacy and safety of caspofungin for treatment of invasive aspergillosis in patients refractory to or intolerant of conventional antifungal therapy. Clin Infect Dis. 2004;39:1563–1571. doi: 10.1086/423381. Epub 2004 Nov 1569. [DOI] [PubMed] [Google Scholar]
  • 84.Marr KA, Boeckh M, Carter RA, Kim HW, Corey L. Combination antifungal therapy for invasive aspergillosis. Clin Infect Dis. 2004;39:797–802. doi: 10.1086/423380. Epub 2004 Aug 2027. [DOI] [PubMed] [Google Scholar]
  • 85.Smith TJ, Khatcheressian J, Lyman GH, Ozer H, Armitage JO, Balducci L, Bennett CL, Cantor SB, Crawford J, Cross SJ, et al. 2006 update of recommendations for the use of white blood cell growth factors: an evidence-based clinical practice guideline. J Clin Oncol. 2006;24:3187–3205. doi: 10.1200/JCO.2006.06.4451. [DOI] [PubMed] [Google Scholar]
  • 86.Price TH, Bowden RA, Boeckh M, Bux J, Nelson K, Liles WC, Dale DC. Phase I/II trial of neutrophil transfusions from donors stimulated with G-CSF and dexamethasone for treatment of patients with infections in hematopoeitic stem cell transplantation. Blood. 2000;95:3302–3309. [PubMed] [Google Scholar]
  • 87.Groll A, Renz S, Gerein V, Schwabe D, Katschan G, Schneider M, Hubner K, Kornhuber B. Fatal haemoptysis associated with invasive pulmonary aspergillosis treated with high-dose amphotericin B and granulocyte-macrophage colony-stimulating factor (GM-CSF) Mycoses. 1992;35:67–75. doi: 10.1111/j.1439-0507.1992.tb00822.x. [DOI] [PubMed] [Google Scholar]
  • *88.Miceli MH, Maertens J, Buve K, Grazziutti M, Woods G, Rahman M, Barlogie B, Anaissie EJ. Immune reconstitution inflammatory syndrome in cancer patients with pulmonary aspergillosis recovering from neutropenia: Proof of principle, description, and clinical and research implications. Cancer. 2007;110:112–120. doi: 10.1002/cncr.22738. [DOI] [PubMed] [Google Scholar]
  • *89.Singh N, Perfect JR. Immune reconstitution syndrome associated with opportunistic mycoses. Lancet Infect Dis. 2007;7:395–401. doi: 10.1016/S1473-3099(07)70085-3. [DOI] [PubMed] [Google Scholar]
  • *90.Safdar A, Rodriguez GH, Lichtiger B, Dickey BF, Kontoyiannis DP, Freireich EJ, Shpall EJ, Raad II, Kantarjian HM, Champlin RE. Recombinant interferon gamma1b immune enhancement in 20 patients with hematologic malignancies and systemic opportunistic infections treated with donor granulocyte transfusions. Cancer. 2006;106:2664–2671. doi: 10.1002/cncr.21929. [DOI] [PubMed] [Google Scholar]
  • **91.Bochud PY, Chien JW, Marr KA, Leisenring WM, Upton A, Janer M, Rodrigues SD, Li S, Hansen JA, Zhao LP, et al. Toll-like receptor 4 polymorphisms and aspergillosis in stem-cell transplantation. N Engl J Med. 2008;359:1766–1777. doi: 10.1056/NEJMoa0802629. [DOI] [PMC free article] [PubMed] [Google Scholar]; This study linked donor toll-like receptor polymorphisms with risk of invasive aspergillosis in allogeneic HSCT recipients.
  • **92.Zaas AK, Liao G, Chien JW, Weinberg C, Shore D, Giles SS, Marr KA, Usuka J, Burch LH, Perera L, et al. Plasminogen alleles influence susceptibility to invasive aspergillosis. PLoS Genet. 2008;4:e1000101. doi: 10.1371/journal.pgen.1000101. [DOI] [PMC free article] [PubMed] [Google Scholar]; This study showed that plasminogen alleles influence susceptibility to invasive aspergillosis in mouse models of invasive aspegillosis an din human HSCT recipients.
  • **93.Fedorova ND, Khaldi N, Joardar VS, Maiti R, Amedeo P, Anderson MJ, Crabtree J, Silva JC, Badger JH, Albarraq A, et al. Genomic islands in the pathogenic filamentous fungus Aspergillus fumigatus. PLoS Genet. 2008;4:e1000046. doi: 10.1371/journal.pgen.1000046. [DOI] [PMC free article] [PubMed] [Google Scholar]; Cross-species comparison between Aspergillus species showed genomic islands that contain substantial genetic variability, and are enriched for pseudogenes, transposons and other repetitive elements; these genetic differences may account for the increased virulence of Aspergillus fumigatus compared to non-pathogenic Aspergillus species in humans.
  • **94.O'Gorman CM, Fuller HT, Dyer PS. Discovery of a sexual cycle in the opportunistic fungal pathogen Aspergillus fumigatus. Nature. 2008;457(7228):471–4. doi: 10.1038/nature07528. [DOI] [PubMed] [Google Scholar]; The discovery of a sexual cycle for A. fumigatus contributes new insight into the biology of this organism, and provides an important tool to study the genetic basis for important traits, such as antifungal drug resistance.

RESOURCES