Invasive fungal diseases (IFDs) represent an important cause of treatment failure in adults with acute leukemia. Because of leukemia’s heterogeneity, the risk for IFDs is highly variable. We therefore apply a risk-adapted antifungal strategy with strong emphasis on pretreatment and day-15 posttreatment to allow earlier and more individualized interventions. We determine pretreatment risks for IFDs based on 4 factors: (1) host fitness for standard therapy (ie, fit, unfit, or frail); (2) leukemia resistance (high vs low probability of achieving complete remission [CR]); (3) anticipated treatment-related toxicity such as neutropenia, mucositis, and steroid-induced immunosuppression; and (4) patient exposure to opportunistic fungi. Accordingly, we stratify patients as high, intermediate, or low risk for IFDs and apply risk-adapted antifungal strategies, including primary or secondary prophylaxis and diagnostic-based preemptive or empiric therapy. Prevention of IFDs also relies on optimizing organ function, decreasing exposure to opportunistic fungi, and improving net state of immunosuppression with use of better-tolerated and investigational agents for unfit patients and those with adverse leukemia biology. Novel targeted and safe therapies that can achieve higher rates of sustained CR among patients with adverse genetics offer the best promise for reducing the burden of IFDs in these patients.

The outcome of adults with acute leukemia (A-Leuk), including acute myelogenous leukemia (AML) and acute lymphocytic leukemia (ALL), has improved over the last decade because of the availability of novel agents and improvements in supportive care such as effective antifungal prophylaxis.1-3 

Despite these advances,1-3  treatment failure remains common and is driven by adverse leukemia genetics (translated into lower probability of achieving complete remission [CR])4-6  and by early treatment-related mortality (e-TRM,7  most commonly caused by infections, particularly invasive fungal diseases [IFDs]), which serve as markers of poor host fitness for therapy.8 

We herein discuss novel risk-adapted and dynamic strategies for the prevention, diagnosis, and treatment of IFDs in patients with A-Leuk, with strong emphasis on pretreatment parameters that allow earlier interventions.

Fungal infections in children and recipients of hematopoietic cell transplantation and those caused by Pneumocystis jiroveci are not discussed. The following clinical course of a patient illustrates some pitfalls in managing IFDs in patients with A-Leuk.

A 45-year-old man was hospitalized for remission-induction of favorable cytogenetics AML using cytarabine and idarubicin (7 + 3). Prophylactic fluconazole was started along with thrice-weekly serum Aspergillus galactomannan index (s-GMI; positive if ≥0.5). On day +5, the patient developed blood culture–negative febrile neutropenia (FN) of 101.5°F with an absolute neutrophil count (ANC) of <100 μL. Piperacillin-tazobactam was started with rapid defervescence. On day +10, low-grade 99.5°F fevers were noted. Physical examination was normal, but s-GMI was elevated at 0.630, rising to 1.735 on day +12. Computed tomography of the chest (chest CT) revealed a 0.4-cm nodule and tree-in-bud pattern infiltrates in the right upper lobe, prompting IV voriconazole for invasive pulmonary aspergillosis (IPA). On day +15, s-GMI reached 2.134, then normalized on day +19 when ANC reached 350/μL. Day +16 bone marrow biopsy showed no blasts. The patient’s clinical course and serial s-GMI remained unremarkable until day +23 when he complained of cough and dyspnea on exertion with hypoxemia. Chest CT revealed marked right upper lobe worsening with wedge-shaped infiltrates. A day +21 sputum yielded Aspergillus versicolor. Because clinical and radiologic worsening coincided with increasing ANC (>4000/μL), but with persistently normal s-GMIs and no other etiology for the infiltrates, a diagnosis of pulmonary inflammatory immune reconstitution syndrome (PIRIS)9  was made, and voriconazole continued. On day +24, he developed early respiratory failure, which rapidly responded to a 3-day course of 2 mg/kg of IV methylprednisolone per day. He was discharged in CR on oral voriconazole until resolution of IPA, with secondary voriconazole prophylaxis throughout consolidation. One year after diagnosis, he remained in sustained CR and free of IPA. This case illustrates several points relevant to IFDs in this patient population, including prevention (fluconazole prophylaxis), early diagnosis (serial s-GMI, cultures, chest CT findings, and confounding presentations such as PIRIS), and treatment (voriconazole), as well as the protective effects of remission status and secondary prophylaxis.

Much has changed in the epidemiology and management of IFDs since our 1995 review in Blood.10 

Increasing mold infections

Invasive candidiasis (IC) was the most frequent IFD in patients with A-Leuk until the introduction of fluconazole,11  which led to a decrease in its incidence, a shift from susceptible (Candida albicans and C tropicalis) to more resistant species (C glabrata and C krusei),12  and an increase in invasive mold infections (IMIs), particularly invasive aspergillosis (IA), followed by fusariosis, mucormycosis, and others.13,14  Prophylaxis with the newly available mold-active triazoles—voriconazole and posaconazole—reduced the incidence of IA but has been associated with breakthrough IMIs with non-Aspergillus molds, particularly mucormycosis.15  In vitro resistance of Aspergillus species to triazoles is reported,16  although its clinical relevance remains unclear.

Geographic and seasonal variations

The epidemiology of IMI is subject to geographic and seasonal variations. For example, scedosporiosis is most frequently observed in Spain and Australia,17,18  whereas the incidence of fusariosis is highest in Houston, Texas,19  and Brazil.14  Seasonal variations in the incidence of aspergillosis and fusariosis were reported in Seattle, Washington,20  and Houston, Texas,19  respectively.

Waterborne IMIs

Patients with A-Leuk may acquire IMI from different sources, including air and water. Aspergillus species, Fusarium species, and the agents of mucormycosis were recovered from air, water, and water-related surfaces of hospitals,21  with demonstration of relatedness between patients’ and waterborne strains.22,23 

The risk for IFDs is higher among AML patients,13  except when ALL-intensified regimens are applied (Table 1). The highest rate is observed following salvage therapy for relapsed-refractory leukemia, although a significant proportion of newly diagnosed patients undergoing remission-induction develop IFDs, most commonly sinus, pulmonary aspergillosis (9% to 15%), or both.24-26  Despite long duration of severe neutropenia (DON; ANC <100/μL, ≥10-14 days), the rate of IA is lower after consolidation therapy (5.7%),27  reflecting the protective effect of CR status.25,26 

Table 1

Risk factors for and risk reduction of IFDs in patients with AML or ALL

Risk factorsRisk reduction
Pretreatment 
Net state of immunosupression 
 Leukemia-related 
  *Lower probability of CR32 
 AML: adverse cytogenetic/gene mutation profiles, WBC ≥50 000/µL, secondary leukemia, MDS or antecedent hematologic disorder >6 mo4,5 
 ALL: adverse cytogenetic/gene mutation profiles, WBC ≥30 000/µL, immunophenotype6  

Assess probability of CR and e-TRM42,43 
Determine risk for IFDs: high, intermediate, or low, and manage accordingly.
Primary mold-active prophylaxis for high-risk patients.
Serial s-GMI tests
Lower cytarabine consolidation for patients with favorable cytogenetics43 
Better-tolerated antileukemic regiments if high risk for e-TRM64-68  
  *Baseline neutropenia
 ANC <500/µL for ≥7 d,26  MDS-related phagocytic dysfunction 

Manage as high risk for IFDs including mold-active prophylaxis
G-CSF for patients with ALL,114  and GM-CSF for those with AML69  
  *Leukemia status35 
 Relapse-refractory > first induction > consolidation 

Manage as high risk for IFDs except for consolidation 
 Treatment-related 
  *Corticosteroids (≥3 wk of >1 mg/kg per d of prednisone equivalent)25,33  Reduce corticosteroid dose, provide antifungal and Pneumocystis prophylaxis 
  *Highly mucotoxic regimen36 
 Cumulative myelotoxicity from rapidly cycling chemotherapy courses 

Fluconazole prophylaxis59  even if low risk for IFDs
Adjust dose density and intensity based on likelihood of CR4243  and e-TRM43-45  
 Host-related 
  *Age >65 y (AML),44  >35 Y (ALL)115 ; Down syndrome

Immunity polymorphisms31 ; pharmacogenomics of antineoplastic drugs117  
Manage as high risk for IFDs, including mold-active prophylaxis
Consider better-tolerated therapies64-68 
Avoid severe drug interactions and monitor clinical toxicity and drug levels31  
Organ dysfunction 
 *High comorbidity scores118  and e-TRM risk43-45  Improve organ function and consider better-tolerated regimens64-68  
 *Chronic obstructive pulmonary disease,38  smoking,39  respiratory viral infection119  Manage as high risk for IFDs, including mold-active antifungal prophylaxis
During influenza season: immunize patient and close contacts vs influenza viruses, prophylaxis with neuraminidase inhibitors, avoid sick visitors,72  smoking cessation 
 *Poor physical functioning
 ECOG/WHO score,120  physiologic status, functional reserve, activities of daily living, gait speed, and others121 
 Hyperglycemia (blood glucose >200 mg/dL for >2 wk)122  

Preemptive physical and occupational therapy

Monitor and correct blood glucose 
Exposure to pathogenic fungi 
 *Prior aspergillosis ± airway colonization by Aspergillus spp41,49 

Room without HEPA filtration73 
Building constructions or renovation74 
Room without water precautions75  
Manage as high risk for IFDs, including mold-active antifungal secondary prophylaxis49,50 
Provide HEPA filtration73 
Ensure safe construction practices74 
Provide water precautions21,75  
Posttreatment 
Net state of immunosupression 
 Neutropenia, severe and prolonged35  (ANC <100/µL for >10 d)
 *Expected severe and prolonged neutropenia
   AML: low CR score,4-6  d 15 blasts >5%,48  no CR by end of induction
   ALL: no CR in 4 wk, persistent MRD32,48 
 Persistent lymphopenia (cells <300 µL) with normal WBC/ANC 
Manage as high risk for IFDs, including mold-active antifungal prophylaxis

Day-15 bone marrow biopsy for blast clearance and MRD48 
G-CSF for patients with ALL114  and G-CSF for those with AML69 
Reduce corticosteroid dose, antifungal and Pneumocystis prophylaxis 
Organ dysfunction 
 *Mucositis36 
 Severe, grade ≥3 for ≥7 d, especially if involving lower gut 

Fluconazole prophylaxis 
Exposure to pathogenic fungi 
 Same as pretreatment
*Multisite colonization by Candida species40 
Central venous catheter123  
Same as pretreatment
Fluconazole prophylaxis, unless a mold-active agent is indicated
Optimal venous catheter care 
Risk factorsRisk reduction
Pretreatment 
Net state of immunosupression 
 Leukemia-related 
  *Lower probability of CR32 
 AML: adverse cytogenetic/gene mutation profiles, WBC ≥50 000/µL, secondary leukemia, MDS or antecedent hematologic disorder >6 mo4,5 
 ALL: adverse cytogenetic/gene mutation profiles, WBC ≥30 000/µL, immunophenotype6  

Assess probability of CR and e-TRM42,43 
Determine risk for IFDs: high, intermediate, or low, and manage accordingly.
Primary mold-active prophylaxis for high-risk patients.
Serial s-GMI tests
Lower cytarabine consolidation for patients with favorable cytogenetics43 
Better-tolerated antileukemic regiments if high risk for e-TRM64-68  
  *Baseline neutropenia
 ANC <500/µL for ≥7 d,26  MDS-related phagocytic dysfunction 

Manage as high risk for IFDs including mold-active prophylaxis
G-CSF for patients with ALL,114  and GM-CSF for those with AML69  
  *Leukemia status35 
 Relapse-refractory > first induction > consolidation 

Manage as high risk for IFDs except for consolidation 
 Treatment-related 
  *Corticosteroids (≥3 wk of >1 mg/kg per d of prednisone equivalent)25,33  Reduce corticosteroid dose, provide antifungal and Pneumocystis prophylaxis 
  *Highly mucotoxic regimen36 
 Cumulative myelotoxicity from rapidly cycling chemotherapy courses 

Fluconazole prophylaxis59  even if low risk for IFDs
Adjust dose density and intensity based on likelihood of CR4243  and e-TRM43-45  
 Host-related 
  *Age >65 y (AML),44  >35 Y (ALL)115 ; Down syndrome

Immunity polymorphisms31 ; pharmacogenomics of antineoplastic drugs117  
Manage as high risk for IFDs, including mold-active prophylaxis
Consider better-tolerated therapies64-68 
Avoid severe drug interactions and monitor clinical toxicity and drug levels31  
Organ dysfunction 
 *High comorbidity scores118  and e-TRM risk43-45  Improve organ function and consider better-tolerated regimens64-68  
 *Chronic obstructive pulmonary disease,38  smoking,39  respiratory viral infection119  Manage as high risk for IFDs, including mold-active antifungal prophylaxis
During influenza season: immunize patient and close contacts vs influenza viruses, prophylaxis with neuraminidase inhibitors, avoid sick visitors,72  smoking cessation 
 *Poor physical functioning
 ECOG/WHO score,120  physiologic status, functional reserve, activities of daily living, gait speed, and others121 
 Hyperglycemia (blood glucose >200 mg/dL for >2 wk)122  

Preemptive physical and occupational therapy

Monitor and correct blood glucose 
Exposure to pathogenic fungi 
 *Prior aspergillosis ± airway colonization by Aspergillus spp41,49 

Room without HEPA filtration73 
Building constructions or renovation74 
Room without water precautions75  
Manage as high risk for IFDs, including mold-active antifungal secondary prophylaxis49,50 
Provide HEPA filtration73 
Ensure safe construction practices74 
Provide water precautions21,75  
Posttreatment 
Net state of immunosupression 
 Neutropenia, severe and prolonged35  (ANC <100/µL for >10 d)
 *Expected severe and prolonged neutropenia
   AML: low CR score,4-6  d 15 blasts >5%,48  no CR by end of induction
   ALL: no CR in 4 wk, persistent MRD32,48 
 Persistent lymphopenia (cells <300 µL) with normal WBC/ANC 
Manage as high risk for IFDs, including mold-active antifungal prophylaxis

Day-15 bone marrow biopsy for blast clearance and MRD48 
G-CSF for patients with ALL114  and G-CSF for those with AML69 
Reduce corticosteroid dose, antifungal and Pneumocystis prophylaxis 
Organ dysfunction 
 *Mucositis36 
 Severe, grade ≥3 for ≥7 d, especially if involving lower gut 

Fluconazole prophylaxis 
Exposure to pathogenic fungi 
 Same as pretreatment
*Multisite colonization by Candida species40 
Central venous catheter123  
Same as pretreatment
Fluconazole prophylaxis, unless a mold-active agent is indicated
Optimal venous catheter care 

Risk stratification is done prior to treatment and on day-15 bone marrow examination. Risk factors are shown according to type: immunosuppression, organ dysfunction, exposure to pathogenic fungi, and to timing (pretreatment or posttreatment). Yeast infections are typically caused by Candida species, whereas aspergillosis is the most common mold infection. ECOG, Eastern Cooperative Oncology Group; G-CSF, granulocyte colony-stimulating factor; GM-CSF, granulocyte macrophage colony-stimulating factor; HEPA, high-efficiency particulate air; MRD, minimal residual disease; WBC, white blood cell; WHO, World Health Organization. *Most important risk factors.

Similarly, patients with ALL develop IFDs during remission induction26,28,29  but may remain at risk after neutrophil recovery because of cumulative treatment-related immunosuppression28  and corticosteroid-induced hyperglycemia.30 

Because of the heterogeneity of A-Leuk, the risk for IFDs is highly variable and results from interactions between net state of immunosuppression, organ dysfunction, and exposure to opportunistic fungi.31 

Immunosuppression is highest among patients with AML and high-risk myelodysplasia (MDS) because of their older age4,5  and adverse cytogenetics (hence, lower CR probability),4-6  neutropenia at diagnosis,5,26  and prolonged treatment-related DON.32  Cumulative corticosteroid doses25,33  and phagocytic dysfunction with antecedent MDS contribute to immunosuppression.34,35 

Organ dysfunction increases IFD risk.4  For example, mucositis following mucotoxic regimens increases the risk for IC36  because Candida species are normal colonizers of gut mucosa,37  whereas preexisting lung pathology, including chronic obstructive pulmonary disease38  and smoking,39  predisposes patients to IA/IMIs following exposure to airborne conidia.25 

Exposure to opportunistic fungi raises the risk for IFDs; for example, multisite colonization by Candida species40  and airway colonization by Aspergillus species41  predispose patients to IC and IPA, respectively.

Pretreatment risk assessment: leukemia genetics as markers for prolonged neutropenia and risk for IFDs

Because delayed diagnosis of IFDs is associated with higher morbidity and mortality, preventive measures should be taken as early as possible, preferably prior to antileukemic therapy.

Prolonged DON determines risk for IFD but because it is unknown prior to therapy, it is of limited value. Identifying pretreatment parameters that predict DON is therefore of paramount importance. Because the definition of CR requires an ANC >1000/μL, failure to enter remission is associated with longer DON and higher rates of severe infections, including IFDs32  and e-TRM.7  Hence, remission status that can be calculated prior to treatment predicts DON,42,43  with the caveat that unfit patients with favorable genetics may not survive long enough to achieve CR.43-45 

Unfavorable prognostic factors for achieving CR and for e-TRM and toxic death include adverse cytogenetics and gene mutation profiles,46  elevated WBC counts,4  older age (arbitrary cutoff point of 65-75 years),44  and poor performance status.44 

We use an online AML model (www.AML-score.org),42  which predicts probability of CR and toxic deaths among newly diagnosed patients (≥60 years old) eligible for intensive therapy.

The importance of pretreatment variables for risk for and outcome of IFDs is illustrated in a study evaluating risk factors for IA among 258 patients with A-Leuk.26  Median DON was 10 days longer among patients with IA than those without (31 vs 21 days, respectively), and DON was a risk factor for IA by univariate but not multivariate analysis. Only high-risk cytogenetics, neutropenia at diagnosis, and prior lung disease were independent predictors for IA. Strikingly, none of 33 patients with favorable cytogenetics developed IA vs 11 of 22 (50%) among those with adverse cytogenetics, and most patients who died of IA-related complications had refractory leukemia.26 

The dominant role of remission status as risk and prognostic factor for IA is further highlighted in a study in which failure to enter CR and older age, but not neutropenia, were independent risk factors for IA following remission-induction for AML,32  and in another study in which progressive leukemia was more predictive of IA-attributable mortality than neutrophil recovery.47 

Additional pretreatment risk factors (the most important shown in bold) are listed in Table 1.

Accordingly, a comprehensive assessment of pretreatment risk factors for infections in general, and IFDs in particular, should be part of routine evaluation in addition to leukemia diagnostics.

Day 15 posttreatment risk assessment

Another predictor of DON is day-15 blast count, as shown in a study of the impact of marrow evaluation for blast clearance: compared with patients with ≥5% day-15 blast count, those with ≤4% had shorter neutropenia (23 vs 33 days; P < .0001), lower rates of bloodstream infections (13.6% vs 23.2%; P = .003), and lower rates of aplastic death (1.8% vs 6.8%; P = .001).48 

Pretreatment assessment of risk for IFDs

Based on factors related to host, leukemia, and fungal exposure, we stratify patients into 3 risk categories for IFDs—high, intermediate, or low—and apply risk-adapted antifungal strategies accordingly.

A) High-risk group: patients with prior aspergillosis;49,50  those on salvage regimens for relapsed-refractory disease; and newly diagnosed patients undergoing remission-induction with any of the following risk factors: neutropenia at baseline,5,26  low CR probability,4-6  age ≥65 years,44  significant pulmonary dysfunction,38  and high e-TRM score4,44  (Table 1).

For these patients, we recommend a mold-active triazole (posaconazole or voriconazole) as primary or secondary prophylaxis, the latter in patients with prior IA or airway colonization with Aspergillus species.41,49 

Antifungal prophylaxis for AML patients undergoing remission-induction therapy was evaluated in randomized controlled trials (RCTs). In one study, posaconazole recipients had significantly lower rates of IFDs (including IA) and a survival benefit compared with those randomized to fluconazole (or itraconazole).51  As a result, posaconazole is increasingly used as prophylaxis.52  Although voriconazole prophylaxis was not tested in a large RCT in the same setting, it is widely used as prophylaxis because of its efficacy in IA.53  Itraconazole is rarely considered for antifungal prophylaxis because of erratic bioavailability.

Problems associated with prophylaxis with mold-active triazoles include toxicity,54  treatment adherence,55  and variable bioavailability,56  in addition to increasing breakthrough IFDs15  and a significant decrease in the sensitivity of the s-GMI, the cornerstone of managing IA.57 

Because of drug-drug interactions between antileukemic agents and mold-active triazoles, it is advisable to start the latter agents 24 hours after the last chemotherapy dose is infused.

In another RCT, prophylaxis with inhaled liposomal amphotericin B (L-AMB) reduced the rate of IPA, although 45% of patients discontinued prophylaxis for at least 1 week.58 

B) Low-risk group: newly diagnosed young patients (≤45 years old) undergoing first remission-induction or consolidation therapy and without risk factors for IFDs. These patients may benefit from fluconazole prophylaxis, particularly if mucotoxicity is expected.36  Serial s-GMI testing is not recommended.

C) Intermediate-risk group: patients not meeting criteria for high- or low-risk groups. A diagnosis-driven preemptive antifungal therapy (DD-AFT) is best suited for these patients, using fluconazole prophylaxis59  with a switch to a mold-active agent after s-GMI seroconversion and compatible clinical and radiologic findings.60 

The DD-AFT was evaluated in an RCT of 240 patients with A-Leuk and recipients of allogeneic hematopoietic cell transplantation randomized to DD-AFT (n = 118) or empiric therapy (n = 122).61  Blood samples for s-GMI and Aspergillus polymerase chain reaction were collected twice-weekly, and results only made available for DD-AFT recipients to allow mold-active therapy for seroconverters. Compared with empiric therapy, DD-AFT reduced the use of antifungal agents (15% vs 32%; P = .002) and increased the rate of IA diagnosis (15% vs 1%; P < .001), with 10 additional cases of IA diagnosed after retrospective analysis of s-GMI and polymerase chain reaction of empirically treated patients.

Several other studies support the DD-AFT strategy.60,62,63 

We recommend DD-AFT because it provides earlier and more frequent diagnosis of IA and prompt therapy for fewer patients, thereby reducing drug cost, toxicity, and drug-drug interactions. Moreover, the diagnostic certainty inherent to an Aspergillus-specific assay is informative for the type and timing of chemotherapy and the need for secondary prophylaxis.50 Figure 1 serves as an example of the DD-AFT management of patients at intermediate-risk for IFDs.

Figure 1

Risk-adapted antifungal strategy. Strategy based on pretreatment and day-15 posttreatment parameters for risk for IFDs in patients with AML undergoing remission-induction therapy. Example shown is for patients at intermediate risk for IFDs.

Figure 1

Risk-adapted antifungal strategy. Strategy based on pretreatment and day-15 posttreatment parameters for risk for IFDs in patients with AML undergoing remission-induction therapy. Example shown is for patients at intermediate risk for IFDs.

Close modal

However, empiric therapy (ie, starting an antifungal agent after 4-7 days of antibiotic-refractory FN) is better suited at institutions at which s-GMIs are not resulted within 48 hours. The agent of choice depends on the antifungal prophylaxis received and may consist of an echinocandin, voriconazole, or L-AMB, the latter being the only option for recipients of mold-active prophylaxis because of infections with triazole-resistant molds, including mucormycosis.15 

Dynamic day-15 posttreatment reassessment for risk for IFDs

Low- and intermediate-risk patients may be reclassified as high-risk for IFDs if their day-15 blast count is ≥5%.48  Prophylactic fluconazole is then replaced by a mold-active agent.

Other measures to prevent IFDs

The following 3 measures can reduce the morbidity and mortality of IFDs (Table 1):

1) Improving net state of immunosuppression by corticosteroid-dose reduction,25,33  intermediate-dose cytarabine consolidation for patients with favorable cytogenetics,43  and application of better-tolerated regimens; for example, arsenic trioxide with all-trans-retinoic acid for acute promyelocytic leukemia,64  tyrosine-kinase inhibitors plus corticosteroids for Philadelphia chromosome–positive ALL,65  first-line hypomethylating agents for elders with AML/MDS,66  and other promising regimens.67,68 

A protective role for GM-CSF against infectious-related toxicities is strongly suggested in an RCT of older AML patients undergoing remission-induction therapy.69 

G-CSF-elicited granulocyte transfusions may serve as a bridge in severely neutropenic patients, in whom neutrophil recovery is not expected within 3 to 4 days.70  Interferon γ, GM-CSF, or both may be useful in nonneutropenic patients, although solid evidence for these strategies is lacking.71 

Lastly, unexpectedly prolonged DON requires evaluation to determine whether it is related to one or more concomitant viral infections, particularly cytomegalovirus, among patients treated for ALL.

2) Optimizing organ function, particularly pulmonary, by preventing respiratory viral infections, avoiding sick visitors,72  and smoking cessation, particularly among patients with chronic obstructive pulmonary disease38  (Table 1).

3) Decreasing exposure to opportunistic fungi by reducing primarily airborne conidia with high-efficiency particulate air filters in patients’ rooms73  and by taking special precautions during any building construction or demolition.74  Reducing secondarily airborne conidia from a water source75  may also be beneficial.

Common manifestations of IFDs during aplasia

The clinical manifestations of IFDs depend on the site of infection, the severity and dynamic nature of immunosuppression, and the infecting pathogen. Hematogenous dissemination with or without metastatic skin lesions is the usual presentation of yeast infections (eg, Candida species), whereas angioinvasion with pneumonia and tissue infarction is the hallmark of mold infections (eg, Aspergillus species). A mixed presentation of hematogenous dissemination and angioinvasion occurs with infections caused by molds with adventitious sporulation; that is, those capable of hematogenous dissemination by yeast-like spores (eg, Fusarium species).31 

During aplasia, unexplained fever is the most common presentation of IFDs, followed by pneumonia with or without sinusitis, and less commonly skin/soft tissue or disseminated infection.

Monitoring patients during aplasia includes history and physical examination, daily blood counts and serum C-reactive protein (CRP), and s-GMI thrice-weekly. Serial CRPs are obtained because of the high negative predictive value (NPV) of normal CRP for severe infections,76  whereas elevated values can suggest an infectious etiology in patients with IFDs who may not develop fever.60  For FN, an infectious disease workup is performed and IV broad-spectrum antibiotics are started. Additional workup is obtained for antibiotic-refractory FN, including chest CT even in the absence of pulmonary findings.76  Patients with negative serum fungal biomarkers but with radiologic findings suggestive of IFD should undergo bronchoalveolar lavage (BAL), with testing of BAL fluids for fungal biomarkers. Table 2 shows additional clinical, laboratory, and imaging findings suggestive of IFDs.

Table 2

Clinical, laboratory, and radiologic findings suggestive of specific fungal diseases in patients with A-Leuk, shown according to presence or absence of myelosuppression

FindingsDuring aplasiaAfter hematopoietic recovery
Hematogenous dissemination: yeasts, mostly Candida species 
 Clinical Hemodynamic instability may develop Abdominal pain and nausea with CDC95  
Skin lesions: painless subcutaneous nodules, maculopapular, pustular, purpura fulminans–like, rarely ecthyma gangrenosum (ulcerative, necrotic) Chorioretinal lesions, joint and bone pain infections
Skin lesions and myalgia less conspicuous than during aplasia 
Rarely myalgia, muscular tenderness  
 Laboratory Positive blood cultures, increased s-BDG86,87  Increased serum alkaline phosphatase in patients with CDC 
Increased s-BDG86,87,90  with persistent infection 
 Imaging Lower yield during aplasia Multiple lesions in liver, spleen, kidneys on CT scan, ultrasound, or MRI in patients with CDC95  
Angioinvasive molds, most commonly Aspergillus species 
 Clinical Pneumonia: dry cough, pleuritic chest pain, pleural rub Pneumonia: respiratory findings may worsen with neutrophil recovery9  
Sinusitis: facial pain, nasal discharge, hard palate ulceration Skin lesions: less conspicuous than during aplasia 
Skin lesions: maculopapular, pustular, or ecthyma gangrenosum31   
 Laboratory Increased s-GMI ± s-BDG82,90 
Positive blood cultures represent contamination31  
Increased s-GMI ± s-BDG86,87  in patients with persistent infection 
 Imaging Chest CT: nodular or wedge-shaped infiltrates or halo sign93  Larger nodules, air crescent sign, cavitation, other94  
Earlier nonspecific findings diagnostic if s-GMI is positive83  Worsening pulmonary infiltrates with rapid neutrophil recovery94  
Angioinvasive molds capable of hematogenous dissemination,31 most commonly Fusarium species 
 Clinical Same as angioinvasive molds, but metastatic skin lesions more common, and target skin lesions may be present (Fusarium species)19  Same as angioinvasive molds, plus chorioretinal lesions, joint and bone pain infections, similar to features of hematogenous dissemination above 
Cellulitis at sites of skin breakdown: Fusarium species19   
Myalgia, muscular tenderness: Fusarium species19   
 Laboratory Same as angioinvasive molds, but blood cultures typically positive Same as angioinvasive molds 
 Imaging Same as angioinvasive molds Same as angioinvasive molds 
FindingsDuring aplasiaAfter hematopoietic recovery
Hematogenous dissemination: yeasts, mostly Candida species 
 Clinical Hemodynamic instability may develop Abdominal pain and nausea with CDC95  
Skin lesions: painless subcutaneous nodules, maculopapular, pustular, purpura fulminans–like, rarely ecthyma gangrenosum (ulcerative, necrotic) Chorioretinal lesions, joint and bone pain infections
Skin lesions and myalgia less conspicuous than during aplasia 
Rarely myalgia, muscular tenderness  
 Laboratory Positive blood cultures, increased s-BDG86,87  Increased serum alkaline phosphatase in patients with CDC 
Increased s-BDG86,87,90  with persistent infection 
 Imaging Lower yield during aplasia Multiple lesions in liver, spleen, kidneys on CT scan, ultrasound, or MRI in patients with CDC95  
Angioinvasive molds, most commonly Aspergillus species 
 Clinical Pneumonia: dry cough, pleuritic chest pain, pleural rub Pneumonia: respiratory findings may worsen with neutrophil recovery9  
Sinusitis: facial pain, nasal discharge, hard palate ulceration Skin lesions: less conspicuous than during aplasia 
Skin lesions: maculopapular, pustular, or ecthyma gangrenosum31   
 Laboratory Increased s-GMI ± s-BDG82,90 
Positive blood cultures represent contamination31  
Increased s-GMI ± s-BDG86,87  in patients with persistent infection 
 Imaging Chest CT: nodular or wedge-shaped infiltrates or halo sign93  Larger nodules, air crescent sign, cavitation, other94  
Earlier nonspecific findings diagnostic if s-GMI is positive83  Worsening pulmonary infiltrates with rapid neutrophil recovery94  
Angioinvasive molds capable of hematogenous dissemination,31 most commonly Fusarium species 
 Clinical Same as angioinvasive molds, but metastatic skin lesions more common, and target skin lesions may be present (Fusarium species)19  Same as angioinvasive molds, plus chorioretinal lesions, joint and bone pain infections, similar to features of hematogenous dissemination above 
Cellulitis at sites of skin breakdown: Fusarium species19   
Myalgia, muscular tenderness: Fusarium species19   
 Laboratory Same as angioinvasive molds, but blood cultures typically positive Same as angioinvasive molds 
 Imaging Same as angioinvasive molds Same as angioinvasive molds 

CDC, chronic disseminated candidiasis; MRI, magnetic resonance imaging.

Diagnostic tools

Blood cultures have moderate sensitivity for hematogenous infections with yeasts77  and molds with adventitious sporulation78  but have no sensitivity for IA.19  Cultures, stains, and histopathology with direct examination of involved sites can be diagnostic (eg, metastatic skin lesions of candidiasis, trichosporonosis,79  and fusariosis).19,78 

Detecting s-GMI and BAL are particularly helpful in diagnosing IA.80,81  The test’s performance in neutropenic patients with A-Leuk is excellent,82  allowing early diagnosis and treatment days before the overt manifestations of IA.60  Early treatment is likely responsible for improved outcomes in IA.83  The kinetics of s-GMI are critical for monitoring response because they correlate with outcome,62,84,85  with rapid and solid response predicted when s-GMI normalizes within 1 week after seroconversion,63  and for distinguishing progressive aspergillosis from other infections or PIRIS.9  The test’s sensitivity, however, is reduced with mold-active prophylaxis.57  The occasional problem of false-positive values is minimized when testing is repeated before infusing broad-spectrum antibiotics. The test is also diagnostic in other IMIs such as fusariosis (sensitivity and specificity of 83% and 67%, respectively).78  We do not consider this result a false positive but a true positive for an IMI, which can be differentiated from IA by its distinguishing clinical and laboratory findings.19 

Detecting circulating serum 1,3-β-D glucan (s-BDG) may be useful in diagnosing various IFDs, including candidiasis, aspergillosis, fusariosis, and others86,87 ; for example, s-BDG was positive in all 10 patients with fusariosis and preceded manifestations by 7 days.88  s-BDG may also be diagnostic for pneumocytosis, a concern in patients with ALL. However, false-positive s-BDG results are common due to several causes86,87  and limit the test’s predictive value, although persistently negative results have a high NPV for IFD.89  The combination of negative s-GMI89  and s-BDG90  practically excludes IFDs, except mucormycosis.82,90 

Chest CT can suggest a diagnosis of IFD,82  although findings vary according to host immunity and may occur with other IMIs91  and other infectious and noninfectious conditions.92  A halo sign93  may suggest early IPA; however, earlier nonspecific findings indicate IPA when serial s-GMI values are elevated.60,83  Larger nodules (>1 cm) may develop with subsequent cavitation with or without air crescent sign after neutrophil recovery.94  A reversed halo sign may suggest mucormycosis.91  Abdominal imaging can detect splenic and hepatic nodules suggestive of chronic disseminated candidiasis,95  and positron emission tomography may be useful in staging extent and response of IFDs but is not recommended.96 

The same principles outlined in “Other measures to prevent IFDs” are also applicable to treatment. Optimizing antifungal therapy is also critical and encompasses selecting the optimal agent, ensuring adequate drug exposure, managing drug-drug interactions, applying objective parameters for outcome assessment, and providing adequate duration of therapy (DOT). Except for hematogenous candidiasis, secondary prophylaxis is required if additional chemotherapy is planned (Table 3).

Table 3

Treatment of IFD in patients with A-Leuk

DiseaseDrug of choiceAlternativeComment
Candidemia Echinocandin: anidulafungin, caspofungin, or micafungin Fluconazole or L-AMB (3 mg/kg per day) Anidulafungin (200-mg loading dose, then 100 mg/d) or caspofungin (70-mg loading dose, then 50 mg/d) or micafungin (100 mg/d). 
Switch to oral agent after a few days when conditions are met. 
Response criteria: clinical findings, blood cultures, CRP. 
DOT: 2 wk. If persistent candidemia, rule out endovascular infection (septic thrombophlebitis, endocarditis); remove central venous catheters; and consider switching to another drug class if no response after the above measures. Continue treatment of 2 wk after resolution of clinical findings and normal CRP. 
Chronic disseminated candidiasis L-AMB (3 mg/kg per day) Echinocandin or fluconazole Switch to oral agent after a few days when conditions are met. 
Immune modulation: consider oral corticosteroids (0.5 mg/kg of prednisone) if symptoms persist despite optimal antifungal therapy.100  
Response criteria: clinical findings, CRP, and s-BDG (if elevated at baseline). Persistence of radiologic lesions is not, by itself, indicative of active infection. 
DOT: continuous throughout consolidation to prevent possible reactivation. 
Aspergillosis Voriconazole53 IV (loading 6 mg/kg every 12 h × 2, then 4 mg/kg every 12 h) L-AMB (3 mg/kg per day) Switch to oral voriconazole after a few days when conditions are met. 
Immune modulation: 
 Decrease dose of immunosuppressive agents, particularly corticosteroids. 
 Consider G-CSF and G-CSF-elicited granulocyte transfusions if neutrophil recovery is not expected within 3-4 d. 
 Consider corticosteroids (prednisone 2 mg/kg per day for 2-3 d if early respiratory failure from PIRIS.9  
Response criteria: clinical findings, s-GMI ± s-BDG if elevated at baseline, CRP. 
Radiologic findings may be misleading (e,. PIRIS9 ) and lag behind response. 
DOT: until recovery from neutropenia, immunosuppression, resolution of clinical findings, and normalization of CRP and s-GMI ± s-BDG (if elevated at baseline). 
Fusariosis L-AMB (3 mg/kg per day) or voriconazole IV at same dose schedule as for aspergillosis ABLC (5 mg/kg per day) Consider adding second agent (voriconazole) if no response after 3-4 d. 
Switch to oral voriconazole when conditions are met. The role of antifungal susceptibility tests in guiding therapy is not known. 
Immune modulation: same as for aspergillosis. 
Response criteria: blood cultures; otherwise, same as for aspergillosis. 
DOT: same as for aspergillosis. 
Mucormycosis L-AMB (5 mg/kg per day) ABLC (5 mg/kg per day or posaconazole IV (loading 300 mg twice a day on day 1, then 300 mg/d) Surgical debridement and control of acidosis if present. 
Switch to oral posaconazole when conditions are met, provided the organism is susceptible to the drug. 
Delayed-release tablets: loading dose 300 mg orally twice a day on day 1, then 300 mg/d 
Oral suspension: 200 mg orally three times a day. 
Immune modulation: same as for fusariosis. 
Response criteria: clinical findings, CRP. 
DOT: until recovery from neutropenia, immunosuppression, resolution of clinical findings, and normalization of CRP. 
DiseaseDrug of choiceAlternativeComment
Candidemia Echinocandin: anidulafungin, caspofungin, or micafungin Fluconazole or L-AMB (3 mg/kg per day) Anidulafungin (200-mg loading dose, then 100 mg/d) or caspofungin (70-mg loading dose, then 50 mg/d) or micafungin (100 mg/d). 
Switch to oral agent after a few days when conditions are met. 
Response criteria: clinical findings, blood cultures, CRP. 
DOT: 2 wk. If persistent candidemia, rule out endovascular infection (septic thrombophlebitis, endocarditis); remove central venous catheters; and consider switching to another drug class if no response after the above measures. Continue treatment of 2 wk after resolution of clinical findings and normal CRP. 
Chronic disseminated candidiasis L-AMB (3 mg/kg per day) Echinocandin or fluconazole Switch to oral agent after a few days when conditions are met. 
Immune modulation: consider oral corticosteroids (0.5 mg/kg of prednisone) if symptoms persist despite optimal antifungal therapy.100  
Response criteria: clinical findings, CRP, and s-BDG (if elevated at baseline). Persistence of radiologic lesions is not, by itself, indicative of active infection. 
DOT: continuous throughout consolidation to prevent possible reactivation. 
Aspergillosis Voriconazole53 IV (loading 6 mg/kg every 12 h × 2, then 4 mg/kg every 12 h) L-AMB (3 mg/kg per day) Switch to oral voriconazole after a few days when conditions are met. 
Immune modulation: 
 Decrease dose of immunosuppressive agents, particularly corticosteroids. 
 Consider G-CSF and G-CSF-elicited granulocyte transfusions if neutrophil recovery is not expected within 3-4 d. 
 Consider corticosteroids (prednisone 2 mg/kg per day for 2-3 d if early respiratory failure from PIRIS.9  
Response criteria: clinical findings, s-GMI ± s-BDG if elevated at baseline, CRP. 
Radiologic findings may be misleading (e,. PIRIS9 ) and lag behind response. 
DOT: until recovery from neutropenia, immunosuppression, resolution of clinical findings, and normalization of CRP and s-GMI ± s-BDG (if elevated at baseline). 
Fusariosis L-AMB (3 mg/kg per day) or voriconazole IV at same dose schedule as for aspergillosis ABLC (5 mg/kg per day) Consider adding second agent (voriconazole) if no response after 3-4 d. 
Switch to oral voriconazole when conditions are met. The role of antifungal susceptibility tests in guiding therapy is not known. 
Immune modulation: same as for aspergillosis. 
Response criteria: blood cultures; otherwise, same as for aspergillosis. 
DOT: same as for aspergillosis. 
Mucormycosis L-AMB (5 mg/kg per day) ABLC (5 mg/kg per day or posaconazole IV (loading 300 mg twice a day on day 1, then 300 mg/d) Surgical debridement and control of acidosis if present. 
Switch to oral posaconazole when conditions are met, provided the organism is susceptible to the drug. 
Delayed-release tablets: loading dose 300 mg orally twice a day on day 1, then 300 mg/d 
Oral suspension: 200 mg orally three times a day. 
Immune modulation: same as for fusariosis. 
Response criteria: clinical findings, CRP. 
DOT: until recovery from neutropenia, immunosuppression, resolution of clinical findings, and normalization of CRP. 

The DOT should be individualized. Switching to oral therapy requires good treatment adherence and intact gut function. Except for hematogenous candidiasis, secondary prophylaxis is required if additional chemotherapy is planned. ABLC, amphotericin B lipid complex.

Selecting the optimal antifungal agent: an individualized approach

We consider several factors when selecting the antifungal agent, including host, pathogen, drug properties, and infection site or sites (Tables 3 and 4). The indications, dosage schedules of antifungal agents, and other treatment measures are shown in Table 3.

Table 4

Practical considerations for individualized selection of antifungal agents for patients with A-Leuk

FactorsSettingAgent of choice, alternatives, and route
Host-related 
 Hemodynamic instability Hematogenous candidiasis Echinocandin; fluconazole and L-AMB as alternatives 
 Organ dysfunction, severe  
  Gastrointestinal tract Mucositis, nausea, vomiting, diarrhea, poor adherence, drug-food interaction IV route 
  Kidneys Tumor lysis syndrome Azoles, echinocandin; avoid amphotericin B products 
  Liver  Echinocandin, L-AMB, ABLC; avoid azoles 
Drug-related 
 Drug-drug interaction Chemotherapy administration Echinocandin, L-AMB, ABLC; avoid mold-active triazoles 
 Drug-food interaction Food intake Echinocandin, L-AMB, fluconazole IV; food intake may alter absorption of azoles 
 Breakthrough infection Infection while on antifungal agent Use different class of antifungal agents 
 Cost and convenience Outpatient setting Oral route always preferable to IV if gut function intact 
Select agent with longest dosing interval 
Infection-related 
 Site of infection Urinary Fluconazole: only agent with urinary concentrations 
Ocular Triazoles, L-AMB; avoid echinocandins (poor distribution) 
CNS Triazoles, L-AMB; avoid echinocandins (poor distribution) 
Pathogen 
Candida species Disseminated, acute and chronic Echinocandin, fluconazole, L-AMB 
  C krusei Disseminated, acute and chronic Echinocandin, L-AMB; avoid fluconazole 
  C glabrata Disseminated, acute and chronic Echinocandin, L-AMB, voriconazole; avoid fluconazole 
  C parapsilosis Disseminated, acute and chronic L-AMB, voriconazole; avoid echinocandins 
Trichosporon spp Disseminated, acute and chronic Fluconazole, other azoles; amphotericin B not effective 
Aspergillus spp Sinus, pulmonary, disseminated Voriconazole, L-AMB, ABLC; no role for fluconazole 
Aspergillus flavus Sinus, pulmonary, disseminated Voriconazole; posaconazole alternative 
Fusarium spp Sinus, pulmonary, cellulitis, disseminated L-AMB, ABLC; voriconazole maintenance if susceptible 
Scedosporium apiospermum Sinus, pulmonary, ocular, CNS, bone and soft tissues, disseminated Voriconazole; posaconazole alternative 
 Black molds Various sites Voriconazole; posaconazole alternative 
 Agents of mucormycosis Sinus, pulmonary, disseminated L-AMB, ABLC; posaconazole maintenance if susceptible 
FactorsSettingAgent of choice, alternatives, and route
Host-related 
 Hemodynamic instability Hematogenous candidiasis Echinocandin; fluconazole and L-AMB as alternatives 
 Organ dysfunction, severe  
  Gastrointestinal tract Mucositis, nausea, vomiting, diarrhea, poor adherence, drug-food interaction IV route 
  Kidneys Tumor lysis syndrome Azoles, echinocandin; avoid amphotericin B products 
  Liver  Echinocandin, L-AMB, ABLC; avoid azoles 
Drug-related 
 Drug-drug interaction Chemotherapy administration Echinocandin, L-AMB, ABLC; avoid mold-active triazoles 
 Drug-food interaction Food intake Echinocandin, L-AMB, fluconazole IV; food intake may alter absorption of azoles 
 Breakthrough infection Infection while on antifungal agent Use different class of antifungal agents 
 Cost and convenience Outpatient setting Oral route always preferable to IV if gut function intact 
Select agent with longest dosing interval 
Infection-related 
 Site of infection Urinary Fluconazole: only agent with urinary concentrations 
Ocular Triazoles, L-AMB; avoid echinocandins (poor distribution) 
CNS Triazoles, L-AMB; avoid echinocandins (poor distribution) 
Pathogen 
Candida species Disseminated, acute and chronic Echinocandin, fluconazole, L-AMB 
  C krusei Disseminated, acute and chronic Echinocandin, L-AMB; avoid fluconazole 
  C glabrata Disseminated, acute and chronic Echinocandin, L-AMB, voriconazole; avoid fluconazole 
  C parapsilosis Disseminated, acute and chronic L-AMB, voriconazole; avoid echinocandins 
Trichosporon spp Disseminated, acute and chronic Fluconazole, other azoles; amphotericin B not effective 
Aspergillus spp Sinus, pulmonary, disseminated Voriconazole, L-AMB, ABLC; no role for fluconazole 
Aspergillus flavus Sinus, pulmonary, disseminated Voriconazole; posaconazole alternative 
Fusarium spp Sinus, pulmonary, cellulitis, disseminated L-AMB, ABLC; voriconazole maintenance if susceptible 
Scedosporium apiospermum Sinus, pulmonary, ocular, CNS, bone and soft tissues, disseminated Voriconazole; posaconazole alternative 
 Black molds Various sites Voriconazole; posaconazole alternative 
 Agents of mucormycosis Sinus, pulmonary, disseminated L-AMB, ABLC; posaconazole maintenance if susceptible 

Antifungal agents include triazoles (fluconazole, voriconazole, posaconazole, and itraconazole), echinocandins (anidulafungin, caspofungin, and micafungin), and amphotericin B and its lipid formulations: L-AMB and ABLC. Severe organ dysfunction refers to grade ≥3 according to common toxicity criteria for adverse events (version 3.0). CNS, central nervous system.

We start IV therapy and switch to an oral agent after improvement, provided treatment adherence is good and gut function is intact (Table 4). We select the oral agent based on recent patient exposure to the same antifungal class, and the agent's expected activity against IFDs. For fungi exhibiting variable susceptibilities (eg, Fusarium species97  to voriconazole, and mucormycosis to posaconazole), we perform antifungal susceptibility testing and switch to these agents accordingly. For immune enhancement, we decrease corticosteroid doses and consider G-CSF and granulocyte transfusions if neutrophil recovery is not expected within 3 to 4 days, as discussed above.

Echinocandins are the drug of choice for hematogenous candidiasis, despite their limited activity against C parapsilosis.98  An alternative is fluconazole, L-AMB, and possibly amphotericin B lipid complex. We discourage the use of deoxycholate amphotericin B because of unacceptable toxicity. We do not routinely remove central venous catheters unless the port or tunnel is infected or if candidemia persists after 5 days of adequate treatment, suggesting an endovascular source and therefore requiring central venous catheter removal and significantly longer DOT99  (Table 3).

Chronic disseminated candidiasis is treated with the same agents used for hematogenous candidiasis. Oral corticosteroids can accelerate clinical improvement if symptoms persist despite antifungal therapy.100  Radiologic persistence of lesions is not, by itself, indicative of active infection.

Voriconazole is the drug of choice for aspergillosis,53  with L-AMB101  as an alternative. A benefit from combination therapy is suggested by the findings of a RCT in which a nonsignificant trend (P = .08) for the primary end point (death at 6 weeks) favored voriconazole plus anidulafungin vs voriconazole plus placebo.102  This limited benefit is likely related to selecting a non-Aspergillus-specific end point.

Worsening pulmonary infiltrates94  coinciding with neutrophil recovery likely represents PIRIS,9  which is distinguished from progressive aspergillosis by a normal s-GMI.63 

The outcome of invasive fusariosis remains largely dependent on persistent neutropenia, corticosteroid-induced immunosuppression, or both.33  Treatment options are shown in Table 3, with little data to support any of these choices. We start L-AMB or voriconazole (Table 3) and add the second drug in the absence of clinical response. After improvement, we switch to oral voriconazole if the organism is susceptible.97 

We treat mucormycosis with L-AMB, debridement of necrotic tissue, and correction of metabolic abnormalities.103  Upon response, we add posaconazole if the organism is susceptible and continue both agents for 1 week to ensure steady-state plasma posaconazole concentrations (PPCs) and then discontinue L-AMB.

Ensuring adequate drug exposure: any role for therapeutic drug monitoring?

The PPCs in patients with A-Leuk are significantly decreased in the presence of mucositis or diarrhea,56,104,105  poor food intake,105  and concomitant receipt of proton pump inhibitors56  or chemotherapeutic agents.105  Lower PPCs have been associated with increased IFDs.104  Compared with oral posaconazole solution, a once-daily tablet significantly improves bioavailability.106  Posaconazole is now available intravenously. Voriconazole is available orally and intravenously and its plasma concentrations vary according to genotype and drug interactions; eg, voriconazole plasma concentrations are significantly decreased with coadministration of glucocorticoids.107 

The role of therapeutic drug monitoring for oral triazoles is unclear because the optimal concentrations remain to be defined and consistently correlated with clinical efficacy. We perform therapeutic drug monitoring when oral triazoles are given as prophylaxis for patients at high-risk for IFD and for the treatment of IFD, when concerns exist about gut function, triazole-related toxicity, or unexplained therapeutic failure.

Managing drug-drug interactions

Different interactions with triazoles must be considered,104  and consultation with a pharmacist is recommended.

1) Interactions in which triazoles affect other drugs,108  requiring monitoring for clinical toxicity and toxic-range blood levels (eg, all-trans-retinoic acid, etoposide, vincristine,109  ifosfamide, and cyclophosphamide).108 

2) Interactions in which other agents such as phenytoin, rifampin, and others decrease or increase triazole exposure (eg, protease inhibitors causing subtherapeutic or supratherapeutic levels of the antifungal agent).

Objective assessment of treatment response

Assessment of IFD response relies on clinical evaluation (fever, metastatic lesions, and others), quantitative laboratory tests (s-GMI, s-BDG, and CRP), and to a lesser extent imaging studies due to the presence of confounding variables and their lag-behind response96  (Table 3).

How long do we treat an IFD?

The DOT is dictated by resolution of the IFD and recovery from neutropenia (ANC >1000/μL), immunosuppression, or both. Evidence-based guidelines for DOT in immunosuppressed nonneutropenic patients are limited,110  and extrapolation from guidelines for HIV-positive patients is suggested.111  When CD4 cells are not measured, we use the absolute lymphocyte count (ALC) to gauge severity of immunosuppression and consider it moderate if ALC is persistently ≤1000/μL, and severe if ALC is persistently ≤300/μL. Ideally, antifungal agents are stopped when the IFD resolves and ALC is consistently ≥1000/μL, absent other causes for lymphocytosis or lymphopenia, such as infections.

A diagnosis of IFD delays chemotherapy and may compromise outcome.49  Hence, careful evaluation of the activity of IFD prior to commencing chemotherapy is critical and relies on objective parameters including fever, s-GMI,63  s-BDG, and CRP. Persistence of some imaging abnormalities does not imply active infection.96  In patients with negative s-GMI, s-BDG, or both at diagnosis, we rely on clinical and radiologic findings and normal CRP values to exclude IFD.

If an IFD is diagnosed, antifungal treatment should be started immediately and chemotherapy delayed until the IFD is controlled, except in rare settings requiring urgent antileukemic intervention. Cytoreduction for elevated peripheral blast count can be achieved with oral hydroxyurea.

Failure to respond to antifungal therapy is not uncommon in patients with A-Leuk112  and is related to persistent profound myelosuppression, immunosuppression, or both. Measures to enhance immunity in “Other measures to prevent IFDs” can be considered. However, every attempt should be made to exclude other causes, including wrong diagnosis (eg, PIRIS),9  mixed fungal and nonfungal infections, suboptimal dose schedule of antifungal agents, and residual effects of IFDs such as persistent neurologic deficits after cerebral infarcts caused by angioinvasive molds. Although acquired drug resistance is rare, primary resistance of Aspergillus species to azoles is an emerging problem in some European countries.113 

IFDs represent an important cause of treatment failure in adults with A-Leuk. Because of leukemia’s heterogeneity, the risk for IFDs is highly variable. We therefore apply a risk-adapted antifungal strategy with strong emphasis on pretreatment and posttreatment variables to allow earlier and more individualized interventions.

Based on pretreatment factors related to host, leukemia, treatment regimen, and fungal exposure, we stratify patients as high, intermediate, or low risk for IFDs and apply measures to prevent and monitor IFDs accordingly. A day-15 blast evaluation allows further risk stratification. Patients at high risk for IFDs who have unfavorable biology or are unfit for therapy can benefit from less-toxic anti-leukemic regimens.64-68 

Novel targeted therapies that can achieve higher rates of sustained CR among patients with adverse genetics offer the best promise for reducing the burden of IFDs in these patients.

M.N. and E.A. wrote and edited the manuscript.

Conflict-of-interest disclosure: M.N. has received honoraria from Merck, Pfizer, Gilead, and Astellas. E.A. has received honoaria from Astellas, Onix, and Millenium.

Correspondence: Elias Anaissie, Hoxworth Center, 3130 Highland Ave, 3rd Floor, Cincinnati, OH, 45219-2316; e-mail: elias114@aol.com or anaissieeliasj@gmail.com.

1
Lichtman
 
MA
A historical perspective on the development of the cytarabine (7days) and daunorubicin (3days) treatment regimen for acute myelogenous leukemia: 2013 the 40th anniversary of 7+3.
Blood Cells Mol Dis
2013
, vol. 
50
 
2
(pg. 
119
-
130
)
2
Pui
 
CH
Evans
 
WE
A 50-year journey to cure childhood acute lymphoblastic leukemia.
Semin Hematol
2013
, vol. 
50
 
3
(pg. 
185
-
196
)
3
Burnett
 
A
Wetzler
 
M
Löwenberg
 
B
Therapeutic advances in acute myeloid leukemia.
J Clin Oncol
2011
, vol. 
29
 
5
(pg. 
487
-
494
)
4
Kantarjian
 
H
O’Brien
 
S
Cortes
 
J
, et al. 
Results of intensive chemotherapy in 998 patients age 65 years or older with acute myeloid leukemia or high-risk myelodysplastic syndrome: predictive prognostic models for outcome.
Cancer
2006
, vol. 
106
 
5
(pg. 
1090
-
1098
)
5
Döhner
 
H
Estey
 
EH
Amadori
 
S
, et al. 
European LeukemiaNet
Diagnosis and management of acute myeloid leukemia in adults: recommendations from an international expert panel, on behalf of the European LeukemiaNet.
Blood
2010
, vol. 
115
 
3
(pg. 
453
-
474
)
6
Chiaretti
 
S
Vitale
 
A
Cazzaniga
 
G
, et al. 
Clinico-biological features of 5202 patients with acute lymphoblastic leukemia enrolled in the Italian AIEOP and GIMEMA protocols and stratified in age cohorts.
Haematologica
2013
, vol. 
98
 
11
(pg. 
1702
-
1710
)
7
Estey
 
EH
Acute myeloid leukemia: 2013 update on risk-stratification and management.
Am J Hematol
2013
, vol. 
88
 
4
(pg. 
318
-
327
)
8
Walter
 
RB
Estey
 
EH
Management of older or unfit patients with acute myeloid leukemia.
Leukemia
2014
 
(Jul 9)
9
Miceli
 
MH
Maertens
 
J
Buvé
 
K
, et al. 
Immune reconstitution inflammatory syndrome in cancer patients with pulmonary aspergillosis recovering from neutropenia: Proof of principle, description, and clinical and research implications.
Cancer
2007
, vol. 
110
 
1
(pg. 
112
-
120
)
10
Uzun
 
O
Anaissie
 
EJ
Antifungal prophylaxis in patients with hematologic malignancies: a reappraisal.
Blood
1995
, vol. 
86
 
6
(pg. 
2063
-
2072
)
11
Bodey
 
G
Bueltmann
 
B
Duguid
 
W
, et al. 
Fungal infections in cancer patients: an international autopsy survey.
Eur J Clin Microbiol Infect Dis
1992
, vol. 
11
 
2
(pg. 
99
-
109
)
12
Abi-Said
 
D
Anaissie
 
E
Uzun
 
O
Raad
 
I
Pinzcowski
 
H
Vartivarian
 
S
The epidemiology of hematogenous candidiasis caused by different Candida species.
Clin Infect Dis
1997
, vol. 
24
 
6
(pg. 
1122
-
1128
)
13
Pagano
 
L
Caira
 
M
Candoni
 
A
, et al. 
The epidemiology of fungal infections in patients with hematologic malignancies: the SEIFEM-2004 study.
Haematologica
2006
, vol. 
91
 
8
(pg. 
1068
-
1075
)
14
Nucci
 
M
Garnica
 
M
Gloria
 
AB
, et al. 
Invasive fungal diseases in haematopoietic cell transplant recipients and in patients with acute myeloid leukaemia or myelodysplasia in Brazil.
Clin Microbiol Infect
2013
, vol. 
19
 
8
(pg. 
745
-
751
)
15
Auberger
 
J
Lass-Flörl
 
C
Aigner
 
M
Clausen
 
J
Gastl
 
G
Nachbaur
 
D
Invasive fungal breakthrough infections, fungal colonization and emergence of resistant strains in high-risk patients receiving antifungal prophylaxis with posaconazole: real-life data from a single-centre institutional retrospective observational study.
J Antimicrob Chemother
2012
, vol. 
67
 
9
(pg. 
2268
-
2273
)
16
van der Linden
 
JW
Camps
 
SM
Kampinga
 
GA
, et al. 
Aspergillosis due to voriconazole highly resistant Aspergillus fumigatus and recovery of genetically related resistant isolates from domiciles.
Clin Infect Dis
2013
, vol. 
57
 
4
(pg. 
513
-
520
)
17
Alastruey-Izquierdo
 
A
Mellado
 
E
Peláez
 
T
, et al. 
FILPOP Study Group
Population-based survey of filamentous fungi and antifungal resistance in Spain (FILPOP Study).
Antimicrob Agents Chemother
2013
, vol. 
57
 
7
(pg. 
3380
-
3387
)
18
Heath
 
CH
Slavin
 
MA
Sorrell
 
TC
, et al. 
Australian Scedosporium Study Group
Population-based surveillance for scedosporiosis in Australia: epidemiology, disease manifestations and emergence of Scedosporium aurantiacum infection.
Clin Microbiol Infect
2009
, vol. 
15
 
7
(pg. 
689
-
693
)
19
Boutati
 
EI
Anaissie
 
EJ
Fusarium, a significant emerging pathogen in patients with hematologic malignancy: ten years’ experience at a cancer center and implications for management.
Blood
1997
, vol. 
90
 
3
(pg. 
999
-
1008
)
20
Panackal
 
AA
Li
 
H
Kontoyiannis
 
DP
, et al. 
Geoclimatic influences on invasive aspergillosis after hematopoietic stem cell transplantation.
Clin Infect Dis
2010
, vol. 
50
 
12
(pg. 
1588
-
1597
)
21
Anaissie
 
EJ
Stratton
 
SL
Dignani
 
MC
, et al. 
Pathogenic molds (including Aspergillus species) in hospital water distribution systems: a 3-year prospective study and clinical implications for patients with hematologic malignancies.
Blood
2003
, vol. 
101
 
7
(pg. 
2542
-
2546
)
22
Anaissie
 
EJ
Kuchar
 
RT
Rex
 
JH
, et al. 
Fusariosis associated with pathogenic fusarium species colonization of a hospital water system: a new paradigm for the epidemiology of opportunistic mold infections.
Clin Infect Dis
2001
, vol. 
33
 
11
(pg. 
1871
-
1878
)
23
Anaissie
 
EJ
Stratton
 
SL
Dignani
 
MC
, et al. 
Pathogenic Aspergillus species recovered from a hospital water system: a 3-year prospective study.
Clin Infect Dis
2002
, vol. 
34
 
6
(pg. 
780
-
789
)
24
Lortholary
 
O
Gangneux
 
JP
Sitbon
 
K
, et al. 
French Mycosis Study Group
Epidemiological trends in invasive aspergillosis in France: the SAIF network (2005-2007).
Clin Microbiol Infect
2011
, vol. 
17
 
12
(pg. 
1882
-
1889
)
25
Walsh
 
TJ
Anaissie
 
EJ
Denning
 
DW
, et al. 
Infectious Diseases Society of America
Treatment of aspergillosis: clinical practice guidelines of the Infectious Diseases Society of America.
Clin Infect Dis
2008
, vol. 
46
 
3
(pg. 
327
-
360
)
26
Chabrol
 
A
Cuzin
 
L
Huguet
 
F
, et al. 
Prophylaxis of invasive aspergillosis with voriconazole or caspofungin during building work in patients with acute leukemia.
Haematologica
2010
, vol. 
95
 
6
(pg. 
996
-
1003
)
27
Cannas
 
G
Pautas
 
C
Raffoux
 
E
, et al. 
Infectious complications in adult acute myeloid leukemia: analysis of the Acute Leukemia French Association-9802 prospective multicenter clinical trial.
Leuk Lymphoma
2012
, vol. 
53
 
6
(pg. 
1068
-
1076
)
28
Henden
 
A
Morris
 
K
Truloff
 
N
Nakagaki
 
M
Kennedy
 
GA
Incidence and outcomes of invasive fungal disease in adult patients with acute lymphoblastic leukemia treated with hyperfractionated cyclophosphamide, vincristine, doxorubicin and dexamethasone: implications for prophylaxis.
Leuk Lymphoma
2013
, vol. 
54
 
6
(pg. 
1329
-
1331
)
29
O’Brien
 
S
Thomas
 
DA
Ravandi
 
F
Faderl
 
S
Pierce
 
S
Kantarjian
 
H
Results of the hyperfractionated cyclophosphamide, vincristine, doxorubicin, and dexamethasone regimen in elderly patients with acute lymphocytic leukemia.
Cancer
2008
, vol. 
113
 
8
(pg. 
2097
-
2101
)
30
Weiser
 
MA
Cabanillas
 
ME
Konopleva
 
M
, et al. 
Relation between the duration of remission and hyperglycemia during induction chemotherapy for acute lymphocytic leukemia with a hyperfractionated cyclophosphamide, vincristine, doxorubicin, and dexamethasone/methotrexate-cytarabine regimen.
Cancer
2004
, vol. 
100
 
6
(pg. 
1179
-
1185
)
31
Anaissie
 
E
Grazziutti
 
M
Nucci
 
M
Anaissie
 
EJ
McGinnis
 
MR
Pfaller
 
MA
Invasive fungal infections in cancer patients.
Clinical Mycology
2009
2nd ed
Philadelphia
Elsevier
(pg. 
431
-
471
)
32
Michallet
 
M
Sobh
 
M
Morisset
 
S
, et al. 
Risk factors for invasive aspergillosis in acute myeloid leukemia patients prophylactically treated with posaconazole.
Med Mycol
2011
, vol. 
49
 
7
(pg. 
681
-
687
)
33
Nucci
 
M
Anaissie
 
EJ
Queiroz-Telles
 
F
, et al. 
Outcome predictors of 84 patients with hematologic malignancies and Fusarium infection.
Cancer
2003
, vol. 
98
 
2
(pg. 
315
-
319
)
34
Fianchi
 
L
Leone
 
G
Posteraro
 
B
, et al. 
Impaired bactericidal and fungicidal activities of neutrophils in patients with myelodysplastic syndrome.
Leuk Res
2012
, vol. 
36
 
3
(pg. 
331
-
333
)
35
van de Peppel
 
RJ
Dekkers
 
OM
von dem Borne
 
PA
de Boer
 
MG
Relapsed and secondary disease drive the risk profile for invasive aspergillosis prior to stem cell transplantation in patients with acute myeloid leukemia or myelodysplastic syndrome.
Med Mycol
2014
, vol. 
52
 
7
(pg. 
699
-
705
)
36
Bow
 
EJ
Meddings
 
JB
Intestinal mucosal dysfunction and infection during remission-induction therapy for acute myeloid leukaemia.
Leukemia
2006
, vol. 
20
 
12
(pg. 
2087
-
2092
)
37
Nucci
 
M
Anaissie
 
E
Revisiting the source of candidemia: skin or gut?
Clin Infect Dis
2001
, vol. 
33
 
12
(pg. 
1959
-
1967
)
38
Guinea
 
J
Torres-Narbona
 
M
Gijón
 
P
, et al. 
Pulmonary aspergillosis in patients with chronic obstructive pulmonary disease: incidence, risk factors, and outcome.
Clin Microbiol Infect
2010
, vol. 
16
 
7
(pg. 
870
-
877
)
39
Verweij
 
PE
Kerremans
 
JJ
Voss
 
A
Meis
 
JF
Fungal contamination of tobacco and marijuana.
JAMA
2000
, vol. 
284
 
22
pg. 
2875
 
40
Martino
 
P
Girmenia
 
C
Micozzi
 
A
De Bernardis
 
F
Boccanera
 
M
Cassone
 
A
Prospective study of Candida colonization, use of empiric amphotericin B and development of invasive mycosis in neutropenic patients.
Eur J Clin Microbiol Infect Dis
1994
, vol. 
13
 
10
(pg. 
797
-
804
)
41
Martino
 
P
Raccah
 
R
Gentile
 
G
Venditti
 
M
Girmenia
 
C
Mandelli
 
F
Aspergillus colonization of the nose and pulmonary aspergillosis in neutropenic patients: a retrospective study.
Haematologica
1989
, vol. 
74
 
3
(pg. 
263
-
265
)
42
Krug
 
U
Röllig
 
C
Koschmieder
 
A
, et al. 
German Acute Myeloid Leukaemia Cooperative Group; Study Alliance Leukemia Investigators
Complete remission and early death after intensive chemotherapy in patients aged 60 years or older with acute myeloid leukaemia: a web-based application for prediction of outcomes.
Lancet
2010
, vol. 
376
 
9757
(pg. 
2000
-
2008
)
43
Ferrara
 
F
Schiffer
 
CA
Acute myeloid leukaemia in adults.
Lancet
2013
, vol. 
381
 
9865
(pg. 
484
-
495
)
44
Walter
 
RB
Othus
 
M
Borthakur
 
G
, et al. 
Prediction of early death after induction therapy for newly diagnosed acute myeloid leukemia with pretreatment risk scores: a novel paradigm for treatment assignment.
J Clin Oncol
2011
, vol. 
29
 
33
(pg. 
4417
-
4423
)
45
Garcia-Garcia
 
FJ
Carcaillon
 
L
Fernandez-Tresguerres
 
J
, et al. 
 
A new operational definition of frailty: the frailty trait scale. J Am Med Dir Assoc. 2014;15(5):371.e7-e371.e13
46
Pastore
 
F
Dufour
 
A
Benthaus
 
T
, et al. 
Combined molecular and clinical prognostic index for relapse and survival in cytogenetically normal acute myeloid leukemia.
J Clin Oncol
2014
, vol. 
32
 
15
(pg. 
1586
-
1594
)
47
Nivoix
 
Y
Velten
 
M
Letscher-Bru
 
V
, et al. 
Factors associated with overall and attributable mortality in invasive aspergillosis.
Clin Infect Dis
2008
, vol. 
47
 
9
(pg. 
1176
-
1184
)
48
Bertoli
 
S
Bories
 
P
Béné
 
MC
, et al. 
Groupe Ouest-Est d’Etude des Leucémies Aiguës et Autres Maladies du Sang (GOELAMS)
Prognostic impact of day 15 blast clearance in risk-adapted remission induction chemotherapy for younger patients with acute myeloid leukemia: long-term results of the multicenter prospective LAM-2001 trial by the GOELAMS study group.
Haematologica
2014
, vol. 
99
 
1
(pg. 
46
-
53
)
49
Even
 
C
Bastuji-Garin
 
S
Hicheri
 
Y
, et al. 
Impact of invasive fungal disease on the chemotherapy schedule and event-free survival in acute leukemia patients who survived fungal disease: a case-control study.
Haematologica
2011
, vol. 
96
 
2
(pg. 
337
-
341
)
50
Cordonnier
 
C
Maury
 
S
Pautas
 
C
, et al. 
Secondary antifungal prophylaxis with voriconazole to adhere to scheduled treatment in leukemic patients and stem cell transplant recipients.
Bone Marrow Transplant
2004
, vol. 
33
 
9
(pg. 
943
-
948
)
51
Cornely
 
OA
Maertens
 
J
Winston
 
DJ
, et al. 
Posaconazole vs. fluconazole or itraconazole prophylaxis in patients with neutropenia.
N Engl J Med
2007
, vol. 
356
 
4
(pg. 
348
-
359
)
52
Vehreschild
 
JJ
Rüping
 
MJ
Wisplinghoff
 
H
, et al. 
Clinical effectiveness of posaconazole prophylaxis in patients with acute myelogenous leukaemia (AML): a 6 year experience of the Cologne AML cohort.
J Antimicrob Chemother
2010
, vol. 
65
 
7
(pg. 
1466
-
1471
)
53
Herbrecht
 
R
Denning
 
DW
Patterson
 
TF
, et al. 
Invasive Fungal Infections Group of the European Organisation for Research and Treatment of Cancer and the Global Aspergillus Study Group
Voriconazole versus amphotericin B for primary therapy of invasive aspergillosis.
N Engl J Med
2002
, vol. 
347
 
6
(pg. 
408
-
415
)
54
Gerber
 
B
Guggenberger
 
R
Fasler
 
D
, et al. 
Reversible skeletal disease and high fluoride serum levels in hematologic patients receiving voriconazole.
Blood
2012
, vol. 
120
 
12
(pg. 
2390
-
2394
)
55
Glasmacher
 
A
Cornely
 
O
Ullmann
 
AJ
, et al. 
Itraconazole Research Group of Germany
An open-label randomized trial comparing itraconazole oral solution with fluconazole oral solution for primary prophylaxis of fungal infections in patients with haematological malignancy and profound neutropenia.
J Antimicrob Chemother
2006
, vol. 
57
 
2
(pg. 
317
-
325
)
56
Vehreschild
 
JJ
Müller
 
C
Farowski
 
F
, et al. 
Factors influencing the pharmacokinetics of prophylactic posaconazole oral suspension in patients with acute myeloid leukemia or myelodysplastic syndrome.
Eur J Clin Pharmacol
2012
, vol. 
68
 
6
(pg. 
987
-
995
)
57
McCulloch
 
E
Ramage
 
G
Rajendran
 
R
, et al. 
Antifungal treatment affects the laboratory diagnosis of invasive aspergillosis.
J Clin Pathol
2012
, vol. 
65
 
1
(pg. 
83
-
86
)
58
Rijnders
 
BJ
Cornelissen
 
JJ
Slobbe
 
L
, et al. 
Aerosolized liposomal amphotericin B for the prevention of invasive pulmonary aspergillosis during prolonged neutropenia: a randomized, placebo-controlled trial.
Clin Infect Dis
2008
, vol. 
46
 
9
(pg. 
1401
-
1408
)
59
Rex
 
JH
Anaissie
 
EJ
Boutati
 
E
Estey
 
E
Kantarjian
 
H
Systemic antifungal prophylaxis reduces invasive fungal in acute myelogenous leukemia: a retrospective review of 833 episodes of neutropenia in 322 adults.
Leukemia
2002
, vol. 
16
 
6
(pg. 
1197
-
1199
)
60
Nucci
 
M
Nouér
 
SA
Grazziutti
 
M
Kumar
 
NS
Barlogie
 
B
Anaissie
 
E
Probable invasive aspergillosis without prespecified radiologic findings: proposal for inclusion of a new category of aspergillosis and implications for studying novel therapies.
Clin Infect Dis
2010
, vol. 
51
 
11
(pg. 
1273
-
1280
)
61
Morrissey
 
CO
Chen
 
SC
Sorrell
 
TC
, et al. 
Australasian Leukaemia Lymphoma Group and the Australia and New Zealand Mycology Interest Group
Galactomannan and PCR versus culture and histology for directing use of antifungal treatment for invasive aspergillosis in high-risk haematology patients: a randomised controlled trial.
Lancet Infect Dis
2013
, vol. 
13
 
6
(pg. 
519
-
528
)
62
Nouér
 
SA
Nucci
 
M
Kumar
 
NS
Grazziutti
 
M
Barlogie
 
B
Anaissie
 
E
Earlier response assessment in invasive aspergillosis based on the kinetics of serum Aspergillus galactomannan: proposal for a new definition.
Clin Infect Dis
2011
, vol. 
53
 
7
(pg. 
671
-
676
)
63
Nouér
 
SA
Nucci
 
M
Kumar
 
NS
Grazziutti
 
M
Restrepo
 
A
Anaissie
 
E
Baseline platelet count and creatinine clearance rate predict the outcome of neutropenia-related invasive aspergillosis.
Clin Infect Dis
2012
, vol. 
54
 
12
(pg. 
e173
-
e183
)
64
Lo-Coco
 
F
Avvisati
 
G
Vignetti
 
M
, et al. 
Gruppo Italiano Malattie Ematologiche dell’Adulto; German-Austrian Acute Myeloid Leukemia Study Group; Study Alliance Leukemia
Retinoic acid and arsenic trioxide for acute promyelocytic leukemia.
N Engl J Med
2013
, vol. 
369
 
2
(pg. 
111
-
121
)
65
Foà
 
R
Vitale
 
A
Vignetti
 
M
, et al. 
GIMEMA Acute Leukemia Working Party
Dasatinib as first-line treatment for adult patients with Philadelphia chromosome-positive acute lymphoblastic leukemia.
Blood
2011
, vol. 
118
 
25
(pg. 
6521
-
6528
)
66
Falantes
 
JF
Calderón
 
C
Márquez-Malaver
 
FJ
, et al. 
Patterns of infection in patients with myelodysplastic syndromes and acute myeloid leukemia receiving azacitidine as salvage therapy. Implications for primary antifungal prophylaxis.
Clin Lymphoma Myeloma Leuk
2014
, vol. 
14
 
1
(pg. 
80
-
86
)
67
Kantarjian
 
HM
O’Brien
 
S
Cortes
 
J
Homoharringtonine/omacetaxine mepesuccinate: the long and winding road to food and drug administration approval.
Clin Lymphoma Myeloma Leuk
2013
, vol. 
13
 
5
(pg. 
530
-
533
)
68
Ghanem
 
H
Kantarjian
 
H
Ohanian
 
M
Jabbour
 
E
The role of clofarabine in acute myeloid leukemia.
Leuk Lymphoma
2013
, vol. 
54
 
4
(pg. 
688
-
698
)
69
Rowe
 
JM
Andersen
 
JW
Mazza
 
JJ
, et al. 
A randomized placebo-controlled phase III study of granulocyte-macrophage colony-stimulating factor in adult patients (> 55 to 70 years of age) with acute myelogenous leukemia: a study of the Eastern Cooperative Oncology Group (E1490).
Blood
1995
, vol. 
86
 
2
(pg. 
457
-
462
)
70
Dignani
 
MC
Anaissie
 
EJ
Hester
 
JP
, et al. 
Treatment of neutropenia-related fungal infections with granulocyte colony-stimulating factor-elicited white blood cell transfusions: a pilot study.
Leukemia
1997
, vol. 
11
 
10
(pg. 
1621
-
1630
)
71
Anaissie
 
EJ
Diagnosis and therapy of fungal infection in patients with leukemia—new drugs and immunotherapy.
Best Pract Res Clin Haematol
2008
, vol. 
21
 
4
(pg. 
683
-
690
)
72
Engelhard
 
D
Mohty
 
B
de la Camara
 
R
Cordonnier
 
C
Ljungman
 
P
European guidelines for prevention and management of influenza in hematopoietic stem cell transplantation and leukemia patients: summary of ECIL-4 (2011), on behalf of ECIL, a joint venture of EBMT, EORTC, ICHS, and ELN.
Transpl Infect Dis
2013
, vol. 
15
 
3
(pg. 
219
-
232
)
73
Abdul Salam
 
ZH
Karlin
 
RB
Ling
 
ML
Yang
 
KS
The impact of portable high-efficiency particulate air filters on the incidence of invasive aspergillosis in a large acute tertiary-care hospital.
Am J Infect Control
2010
, vol. 
38
 
4
(pg. 
e1
-
e7
)
74
Haiduven
 
D
Nosocomial aspergillosis and building construction.
Med Mycol
2009
, vol. 
47
 
Suppl 1
(pg. 
S210
-
S216
)
75
Anaissie
 
EJ
Stratton
 
SL
Dignani
 
MC
, et al. 
Cleaning patient shower facilities: a novel approach to reducing patient exposure to aerosolized Aspergillus species and other opportunistic molds.
Clin Infect Dis
2002
, vol. 
35
 
8
(pg. 
E86
-
E88
)
76
Prat
 
C
Sancho
 
JM
Dominguez
 
J
, et al. 
Evaluation of procalcitonin, neopterin, C-reactive protein, IL-6 and IL-8 as a diagnostic marker of infection in patients with febrile neutropenia.
Leuk Lymphoma
2008
, vol. 
49
 
9
(pg. 
1752
-
1761
)
77
Kami
 
M
Machida
 
U
Okuzumi
 
K
, et al. 
Effect of fluconazole prophylaxis on fungal blood cultures: an autopsy-based study involving 720 patients with haematological malignancy.
Br J Haematol
2002
, vol. 
117
 
1
(pg. 
40
-
46
)
78
Nucci
 
M
Carlesse
 
F
Cappellano
 
P
, et al. 
Earlier diagnosis of invasive fusariosis with Aspergillus serum galactomannan testing.
PLoS ONE
2014
, vol. 
9
 
1
pg. 
e87784
 
79
Anaissie
 
E
Gokaslan
 
A
Hachem
 
R
, et al. 
Azole therapy for trichosporonosis: clinical evaluation of eight patients, experimental therapy for murine infection, and review.
Clin Infect Dis
1992
, vol. 
15
 
5
(pg. 
781
-
787
)
80
Maertens
 
J
Verhaegen
 
J
Demuynck
 
H
, et al. 
Autopsy-controlled prospective evaluation of serial screening for circulating galactomannan by a sandwich enzyme-linked immunosorbent assay for hematological patients at risk for invasive Aspergillosis.
J Clin Microbiol
1999
, vol. 
37
 
10
(pg. 
3223
-
3228
)
81
Maertens
 
J
Maertens
 
V
Theunissen
 
K
, et al. 
Bronchoalveolar lavage fluid galactomannan for the diagnosis of invasive pulmonary aspergillosis in patients with hematologic diseases.
Clin Infect Dis
2009
, vol. 
49
 
11
(pg. 
1688
-
1693
)
82
Maertens
 
J
Theunissen
 
K
Verhoef
 
G
, et al. 
Galactomannan and computed tomography-based preemptive antifungal therapy in neutropenic patients at high risk for invasive fungal infection: a prospective feasibility study.
Clin Infect Dis
2005
, vol. 
41
 
9
(pg. 
1242
-
1250
)
83
Nucci
 
M
Nouér
 
SA
Cappone
 
D
Anaissie
 
E
Early diagnosis of invasive pulmonary aspergillosis in hematologic patients: an opportunity to improve the outcome.
Haematologica
2013
, vol. 
98
 
11
(pg. 
1657
-
1660
)
84
Miceli
 
MH
Grazziutti
 
ML
Woods
 
G
, et al. 
Strong correlation between serum aspergillus galactomannan index and outcome of aspergillosis in patients with hematological cancer: clinical and research implications.
Clin Infect Dis
2008
, vol. 
46
 
9
(pg. 
1412
-
1422
)
85
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
, vol. 
110
 
4
(pg. 
830
-
834
)
86
Lamoth
 
F
Cruciani
 
M
Mengoli
 
C
, et al. 
Third European Conference on Infections in Leukemia (ECIL-3)
β-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
, vol. 
54
 
5
(pg. 
633
-
643
)
87
Sulahian
 
A
Porcher
 
R
Bergeron
 
A
, et al. 
Use and limits of (1-3)-β-d-glucan assay (Fungitell), compared to galactomannan determination (Platelia Aspergillus), for diagnosis of invasive aspergillosis.
J Clin Microbiol
2014
, vol. 
52
 
7
(pg. 
2328
-
2333
)
88
Nucci
 
M
Garnica
 
M
Reis
 
H
, et al. 
 
Performance of 1,3-beta-D-glucan (BDG) in the diagnosis and monitoring of invasive fusariosis [abstract]. Paper presented at the 23rd European Congress of Clinical Microbiology and Infectious Diseases. April 27-30, 2013. Berlin, Germany. Abstract O-166
89
Marchetti
 
O
Lamoth
 
F
Mikulska
 
M
Viscoli
 
C
Verweij
 
P
Bretagne
 
S
European Conference on Infections in Leukemia (ECIL) Laboratory Working Groups
ECIL recommendations for the use of biological markers for the diagnosis of invasive fungal diseases in leukemic patients and hematopoietic SCT recipients.
Bone Marrow Transplant
2012
, vol. 
47
 
6
(pg. 
846
-
854
)
90
Theel
 
ES
Doern
 
CD
β-D-glucan testing is important for diagnosis of invasive fungal infections.
J Clin Microbiol
2013
, vol. 
51
 
11
(pg. 
3478
-
3483
)
91
Legouge
 
C
Caillot
 
D
Chrétien
 
ML
, et al. 
The reversed halo sign: pathognomonic pattern of pulmonary mucormycosis in leukemic patients with neutropenia?
Clin Infect Dis
2014
, vol. 
58
 
5
(pg. 
672
-
678
)
92
Escuissato
 
DL
Gasparetto
 
EL
Marchiori
 
E
, et al. 
Pulmonary infections after bone marrow transplantation: high-resolution CT findings in 111 patients.
AJR Am J Roentgenol
2005
, vol. 
185
 
3
(pg. 
608
-
615
)
93
Caillot
 
D
Casasnovas
 
O
Bernard
 
A
, et al. 
Improved management of invasive pulmonary aspergillosis in neutropenic patients using early thoracic computed tomographic scan and surgery.
J Clin Oncol
1997
, vol. 
15
 
1
(pg. 
139
-
147
)
94
Caillot
 
D
Couaillier
 
JF
Bernard
 
A
, et al. 
Increasing volume and changing characteristics of invasive pulmonary aspergillosis on sequential thoracic computed tomography scans in patients with neutropenia.
J Clin Oncol
2001
, vol. 
19
 
1
(pg. 
253
-
259
)
95
Anaissie
 
E
Bodey
 
GP
Kantarjian
 
H
, et al. 
Fluconazole therapy for chronic disseminated candidiasis in patients with leukemia and prior amphotericin B therapy.
Am J Med
1991
, vol. 
91
 
2
(pg. 
142
-
150
)
96
Mahfouz
 
T
Miceli
 
MH
Saghafifar
 
F
, et al. 
18F-fluorodeoxyglucose positron emission tomography contributes to the diagnosis and management of infections in patients with multiple myeloma: a study of 165 infectious episodes.
J Clin Oncol
2005
, vol. 
23
 
31
(pg. 
7857
-
7863
)
97
Debourgogne
 
A
de Hoog
 
S
Lozniewski
 
A
Machouart
 
M
Amphotericin B and voriconazole susceptibility profiles for the Fusarium solani species complex: comparison between the E-test and CLSI M38-A2 microdilution methodology.
Eur J Clin Microbiol Infect Dis
2012
, vol. 
31
 
4
(pg. 
615
-
618
)
98
Ruhnke
 
M
Paiva
 
JA
Meersseman
 
W
, et al. 
Anidulafungin for the treatment of candidaemia/invasive candidiasis in selected critically ill patients.
Clin Microbiol Infect
2012
, vol. 
18
 
7
(pg. 
680
-
687
)
99
Nucci
 
M
Anaissie
 
E
Betts
 
RF
, et al. 
Early removal of central venous catheter in patients with candidemia does not improve outcome: analysis of 842 patients from 2 randomized clinical trials.
Clin Infect Dis
2010
, vol. 
51
 
3
(pg. 
295
-
303
)
100
Legrand
 
F
Lecuit
 
M
Dupont
 
B
, et al. 
Adjuvant corticosteroid therapy for chronic disseminated candidiasis.
Clin Infect Dis
2008
, vol. 
46
 
5
(pg. 
696
-
702
)
101
Cornely
 
OA
Maertens
 
J
Bresnik
 
M
, et al. 
AmBiLoad Trial Study Group
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
, vol. 
44
 
10
(pg. 
1289
-
1297
)
102
Marr
 
KA
Schlamm
 
H
Rottinghaus
 
S
, et al. 
A randomised, double-blind study of combination antifungal therapy with voriconazole and anidulafungin versus voriconazole monotherapy for primary treatment of invasive aspergillosis [abstract].
Clin Microbiol Infect
2012
, vol. 
18
 pg. 
713
 
103
Skiada
 
A
Lanternier
 
F
Groll
 
AH
, et al. 
European Conference on Infections in Leukemia
Diagnosis and treatment of mucormycosis in patients with hematological malignancies: guidelines from the 3rd European Conference on Infections in Leukemia (ECIL 3).
Haematologica
2013
, vol. 
98
 
4
(pg. 
492
-
504
)
104
Cojutti
 
P
Candoni
 
A
Simeone
 
E
Franceschi
 
L
Fanin
 
R
Pea
 
F
Antifungal prophylaxis with posaconazole in patients with acute myeloid leukemia: dose intensification coupled with avoidance of proton pump inhibitors is beneficial in shortening time to effective concentrations.
Antimicrob Agents Chemother
2013
, vol. 
57
 
12
(pg. 
6081
-
6084
)
105
Vaes
 
M
Hites
 
M
Cotton
 
F
, et al. 
Therapeutic drug monitoring of posaconazole in patients with acute myeloid leukemia or myelodysplastic syndrome.
Antimicrob Agents Chemother
2012
, vol. 
56
 
12
(pg. 
6298
-
6303
)
106
Krishna
 
G
Ma
 
L
Martinho
 
M
Preston
 
RA
O’Mara
 
E
A new solid oral tablet formulation of posaconazole: a randomized clinical trial to investigate rising single- and multiple-dose pharmacokinetics and safety in healthy volunteers.
J Antimicrob Chemother
2012
, vol. 
67
 
11
(pg. 
2725
-
2730
)
107
Dolton
 
MJ
Ray
 
JE
Chen
 
SC
Ng
 
K
Pont
 
LG
McLachlan
 
AJ
Multicenter study of voriconazole pharmacokinetics and therapeutic drug monitoring.
Antimicrob Agents Chemother
2012
, vol. 
56
 
9
(pg. 
4793
-
4799
)
108
Gubbins
 
P
Anaissie
 
EJ
Anaissie
 
EJ
McGinnis
 
MR
Pfaller
 
MA
Antifungal therapy.
Clinical Mycology
2009
2nd ed
Philadelphia
Churchill Livingstone
(pg. 
161
-
195
)
109
Harnicar
 
S
Adel
 
N
Jurcic
 
J
Modification of vincristine dosing during concomitant azole therapy in adult acute lymphoblastic leukemia patients.
J Oncol Pharm Pract
2009
, vol. 
15
 
3
(pg. 
175
-
182
)
110
Anaissie
 
EJ
Kontoyiannis
 
DP
O’Brien
 
S
, et al. 
Infections in patients with chronic lymphocytic leukemia treated with fludarabine.
Ann Intern Med
1998
, vol. 
129
 
7
(pg. 
559
-
566
)
111
Aberg
 
JA
Gallant
 
JE
Ghanem
 
KG
Emmanuel
 
P
Zingman
 
BS
Horberg
 
MA
Infectious Diseases Society of America
Primary care guidelines for the management of persons infected with HIV: 2013 update by the HIV Medicine Association of the Infectious Diseases Society of America.
Clin Infect Dis
2014
, vol. 
58
 
1
(pg. 
1
-
10
)
112
Nucci
 
M
Perfect
 
JR
When primary antifungal therapy fails.
Clin Infect Dis
2008
, vol. 
46
 
9
(pg. 
1426
-
1433
)
113
van der Linden
 
JW
Snelders
 
E
Kampinga
 
GA
, et al. 
Clinical implications of azole resistance in Aspergillus fumigatus, The Netherlands, 2007-2009.
Emerg Infect Dis
2011
, vol. 
17
 
10
(pg. 
1846
-
1854
)
114
Giebel
 
S
Thomas
 
X
Hallbook
 
H
, et al. 
The prophylactic use of granulocyte-colony stimulating factor during remission induction is associated with increased leukaemia-free survival of adults with acute lymphoblastic leukaemia: a joint analysis of five randomised trials on behalf of the EWALL.
Eur J Cancer
2012
, vol. 
48
 
3
(pg. 
360
-
367
)
115
Ravandi
 
F
Faderl
 
S
Kebriaei
 
P
Kantarjian
 
H
Modern treatment programs for adults with acute lymphoblastic leukemia.
Curr Hematol Malig Rep
2007
, vol. 
2
 
3
(pg. 
169
-
175
)
116
Patrick
 
K
Wade
 
R
Goulden
 
N
, et al. 
Outcome of Down syndrome associated acute lymphoblastic leukaemia treated on a contemporary protocol.
Br J Haematol
2014
, vol. 
165
 
4
(pg. 
552
-
555
)
117
Emadi
 
A
Karp
 
JE
The clinically relevant pharmacogenomic changes in acute myelogenous leukemia.
Pharmacogenomics
2012
, vol. 
13
 
11
(pg. 
1257
-
1269
)
118
Meyers
 
J
Yu
 
Y
Kaye
 
JA
Davis
 
KL
Medicare fee-for-service enrollees with primary acute myeloid leukemia: an analysis of treatment patterns, survival, and healthcare resource utilization and costs.
Appl Health Econ Health Policy
2013
, vol. 
11
 
3
(pg. 
275
-
286
)
119
Garcia-Vidal
 
C
Barba
 
P
Arnan
 
M
, et al. 
Invasive aspergillosis complicating pandemic influenza A (H1N1) infection in severely immunocompromised patients.
Clin Infect Dis
2011
, vol. 
53
 
6
(pg. 
e16
-
e19
)
120
Sørensen
 
JB
Klee
 
M
Palshof
 
T
Hansen
 
HH
Performance status assessment in cancer patients. An inter-observer variability study.
Br J Cancer
1993
, vol. 
67
 
4
(pg. 
773
-
775
)
121
Klepin
 
HD
Geiger
 
AM
Tooze
 
JA
, et al. 
Geriatric assessment predicts survival for older adults receiving induction chemotherapy for acute myelogenous leukemia.
Blood
2013
, vol. 
121
 
21
(pg. 
4287
-
4294
)
122
Dare
 
JM
Moppett
 
JP
Shield
 
JP
Hunt
 
LP
Stevens
 
MC
The impact of hyperglycemia on risk of infection and early death during induction therapy for acute lymphoblastic leukemia (ALL).
Pediatr Blood Cancer
2013
, vol. 
60
 
12
(pg. 
E157
-
E159
)
123
Gamaletsou
 
MN
Walsh
 
TJ
Zaoutis
 
T
, et al. 
A prospective, cohort, multicentre study of candidaemia in hospitalized adult patients with haematological malignancies.
Clin Microbiol Infect
2014
, vol. 
20
 
1
(pg. 
O50
-
O57
)
Sign in via your Institution