Improvements in anticancer treatments, the ability to modify myelosuppression profiles, greater duration and intensity of immunosuppression, and the variety of available antimicrobial therapies have influenced the spectrum of pathogens associated with invasive fungal infection complicating treatment of hematological malignancies and hematopoietic stem cell transplantation. The approaches to the management of these infections encompass strategies of prevention for all those at risk, pre-emptive therapy based upon surrogates of infection before the onset of clinical disease, empirical therapy for patients with clinical evidence of early disease, and directed or targeted therapy for infected patients with established disease. Chemoprophylaxis is effective if applied to the highest risk patients over the duration of the risk. Pre-emptive strategies, while promising, have yet to be validated and linked to reliably predictive nonmicrobiological diagnostic techniques. Empirical antifungal therapy, as it is currently applied, now seems questionable. Patients with probable or proven invasive fungal infection still have suboptimal outcomes despite the availability of promising anti-fungal agents. Strategies examining the concept of dose-intensity and combination regimens require careful study and cannot yet be regarded as an acceptable standard of practice.

Interest in the field of medical mycology has expanded considerably as the numbers of immuno- and myelosuppressed patients susceptible to opportunistic invasive fungal infections (IFI) increase. While the majority (95–97%) of invasive yeast infections are due to five species, predominantly Candida albicans, followed by C. tropicalis, C. glabrata, C. parapsilosis, and C. krusei, emerging importance of infections due to non-albicans Candida spp. including C. glabrata, C. parapsilosis, and C. krusei in high-risk cancer patients is noteworthy.1 The remaining 3% to 5% of pathogens include other non-albicans Candida spp.. and non-Candida yeasts such as Trichosporon spp., Cryptococcus spp., Blastoschizomyces spp., and Malassezia spp..2,3 The risks for IFI among hematopoietic stem cell transplants (HSCT) increases as the disparity of the donor:recipient pair (autologous HSCT, 0.6%; matched related donor HSCT, 3.7%; mismatched related or unrelated donor HSCT, 5.9%, P < 0.0001).4 The changing character of the HSCT populations together with effective anti-Candida agents contribute to a changing spectrum of opportunistic mycoses. In a comparison of transplant patients with IFI to a more general population of patients at risk, the proportion of Candida infections decreased by approximately 45% (77% to 42%); however, that due to Aspergillus spp. has more than doubled (13% to 29%) and that due to other environmental moulds including Fusarium spp., Scedosporium spp., the zygomycetes, and the less common dematiacious fungi, Trichoderma spp., Paecilomyces spp., and Scopulariopsis spp., more than tripled (4% to 14%).4,5 

First-order risk groups for IFI include those with underlying conditions such as cancer or myelosuppression, those undergoing abdominal surgery, prematurity, and advanced age. Additional second-order risk factors applicable to these groups include indwelling vascular catheters, broad-spectrum antibacterial therapy, renal insufficiency, fungal colonization, prolonged ICU admissions, and parenteral nutrition.1 Among HSCT recipients, additional risk factors include conditioning regimen (myeloablative versus non-myeloablative); source of stem cells; donor:recipient disparity; graft-versus-host disease; augmented combination immunosuppression with agents such as calcinurin inhibitors, mycophenolate, corticosteroids and infliximab; prolonged severe neutropenia; and environmental exposures.

Even though the event rates for these infections are low by comparison to bacterial diseases, the mortality rates are disproportionately high. Higher clinical indices of suspicion, improved diagnostic imaging techniques, and molecular diagnostic strategies have improved our ability to identify patients at risk for these diseases.3,6,8 

Diagnostic criteria have been developed to provide consistency in the conduct of clinical trials of antifungal therapy9; however, clinicians have been discouraged from using these criteria in clinical circumstances.10 The availability of newer antifungal agents with differing mechanisms of action has encouraged their application to a spectrum of clinical circumstances. This review is intended to highlight some aspects of treatment of invasive fungal infection (IFI) important to the management of patients with hematological malignancies and stem cell transplant (SCT) and to expand upon several previous excellent reviews presented at the American Society of Hematology.11,12 

An individual patient may be eligible for one or more of these approaches at different times during the trajectory of the course of treatment for the underlying disease. Prophylactic therapy targets a population of patients at risk for IFI over a defined period of risk, but who do not yet have evidence of either infection or clinical disease. Pre-emptive or presumptive therapy targets a population of patients with evidence of IFI based upon a surrogate marker such as an antigen or genomic detection test, but without evidence of clinical disease. Empiric antifungal therapy, on the other hand, may be regarded as a variant of pre-emptive therapy where persistent neutropenic fever despite broad-spectrum antibacterial therapy is considered the surrogate marker of IFI. Directed or targeted therapy applies to patients with evidence of infection and clinical disease. It remains unclear whether the array of available imaging and molecular diagnostic techniques should be applied for screening purposes to identify high-risk patients eligible for pre-emptive therapy or for non-culture–based diagnosis in the presence of signs and symptoms suggestive of invasive fungal infection.

What are the current chemoprophylaxis practices?

Table 1  details the proportions of centers in the US, Japan, and Europe reporting the administration of antifungal prophylaxis with fluconazole or extended-spectrum mould-active azoles in high-risk patients between 2001 and 2005. Most HSCT centers in these regions report using antifungal chemoprophylaxis, fluconazole still being the most commonly prescribed. Of note, however, the higher proportion of European centers reporting the administration of mould-active azoles in 2005 reflects the growing concern for invasive mould infections.

Fluconazole and itraconazole have been administered over a range of oral doses (50–400 mg and up to 10 mg/kg/d, respectively); however, evidence suggests that among highest risk patients higher doses are more effective.13,14 The ability to switch from the oral to an intravenous formulation for the same antifungal product in the setting of severe oral mucositis is an advantage. Prophylactic voriconazole in allogeneic HSCT recipients (oral or IV 200 mg BID) is currently under investigation. Two large randomized, fluconazole- or itraconazole-based controlled clinical trials in acute leukemia patients15 and in HSCT recipients with graft-versus-host disease16 have demonstrated the efficacy of oral posaconazole (200 mg thrice daily) for reducing invasive mould infections. Prophylactic micafungin17 and caspofungin18 have been studied at daily IV doses of 50 mg.

When should antifungal prophylaxis begin and end?

Table 2  shows the distribution of reported start and end dates for antifungal prophylaxis reported in the literature.13,15,16,19,23 Prophylaxis should usually be initiated in parallel with the administration of cytotoxic therapy in order to ensure a protective effect at the time of maximal neutropenia and intestinal epithelial damage. Concerns over drug interactions have compelled some investigators to modify the application of triazoles-based prophylaxis until after the administration of cytotoxic therapy.15,16,21 The end date should be dictated by the termination of the specific risk. Mould-active prophylaxis may require administration into the late post-engraftment period in allogeneic HSCT for those patients with higher risk due to acute or chronic graft-versus-host disease requiring augmented immunosuppressive therapy.16,21 

How effective are these strategies?

Several systematic reviews evaluating the published clinical trials of antifungal prophylaxis efficacy are available for review.24 Moreover, two recent large randomized controlled trials have been published in abstract form15,16 demonstrating treatment effects for a variety of clinically important outcomes, including use of empirical antifungal therapy for persistent neutropenic fever, mucosal colonization, superficial fungal infection, proven IFI, IFI-related mortality, and all-cause mortality (particularly in subsets of patients with prolonged severe neutropenia13).

The interest in this strategy is based upon the observation that early detection is associated with better outcomes. A multicenter randomized German study compared preemptive and empirical antifungal therapy with liposomal amphotericin B among allogeneic HSCT recipients receiving fluconazole prophylaxis25 based on serial serum PCR fungal DNA detection studies. The pre-emptive group received more antifungal therapy than the empirical therapy group (109 of 196, 56%, vs 76 of 207, 37%, respectively, P < 0.001). Although the numbers of documented IFI were similar (5.6% versus 7.7%, P = 0.396, respectively), the 30-day mortality was reduced by 67% in the pre-emptive group (2.0% versus 6.3%, P = 0.034).

A second study in acute leukemia patients receiving fluconazole prophylaxis examined an algorithm-based preemptive approach based upon serial diagnostic testing and clinical monitoring.26 Only patients with ≥ 2 positive serum galactomannan assays or CT and or bronchoscopic evidence for mould infection received antifungal therapy. A total of 41 of 117 febrile neutropenic episodes (35%) had persistent neutropenic fever; however, only 9 patients (22% of the 41 persistent neutropenic fevers; 8% of the original febrile neutropenic episodes) satisfied the pre-defined criteria for antifungal therapy—a 78% relative risk reduction in antifungal therapy use.

Despite these promising observations, the appearance of a clinical or radiological marker such as a suggestive nodular pulmonary infiltrate on computerized thoracic tomography in a high-risk patient will compel the anxious physician to initiate antimould therapy independent of molecular markers. Pending validation in larger randomized studies, these important studies not only argue the feasibility of the pre-emptive approach but also challenge the empirical approach as a standard of practice.

During the early 1980s one quarter to one third of severely neutropenic cancer patients with persistent or recrudescent fever despite receiving broad-spectrum antibacterial therapy developed IFI. Empirical amphotericin B deoxycholate reduced the incidence of IFI and overall mortality by 50%–80% and 23%–45%, respectively. The early seminal reports have become the rationale for the current standard of practice despite the lack of statistical robustness in the original observations for defervescence and survival (P = 0.08 and P = 0.23, respectively).27 

Fever is a poorly predictive surrogate upon which to base an intervention such as empirical antifungal therapy.28 Between 35% to 69% of leukemia patients and 56% to 82% of HSCT receive empirical antifungal therapy, yet proven IFI occurs in only 2% to 15%,13,15,16,29 suggesting that the current guidelines30 based upon persistent neutropenic fever are significantly flawed and may result in unjustifiable excess treatment-related toxicities and resource expenditures.28 

Invasive Candidiasis

Myeloid recovery is an independent predictor of outcome among patients with Candida spp. bloodstream infections. Very few studies of invasive candidiasis (IC), particularly bloodstream infections, include data on neutropenic patients.

The results of individual trials have suggested that fluconazole at daily doses of 400 milligrams and amphotericin B deoxycholate at daily doses of 0.5 to 0.6 milligrams per kilogram are similarly effective in non-neutropenic patients; however, a pooled analysis of randomized-controlled trials comparing the efficacy of fluconazole and amphotericin B deoxycholate for IC suggests the possible superiority of amphotericin B (62% of 345 patients versus 70% of 343 patients, χ2 = 5.206, P = 0.023, OR 1.47, 95%CI 1.07 to 2.03) (Figure 1 ),31,34 despite the greater amphotericin B-related renal toxicity noted in previous pooled analyses (OR 3.20, 95%CI 1.61 to 5.01).35 

Voriconazole, an extended-spectrum azole, offers a safe and effective alternative to amphotericin B-based regimens for the treatment of IC36( Figure 1 ). Itraconazole has been recommended for maintenance treatment and secondary prophylaxis of invasive aspergillosis, the primary treatment of dimorphic fungal infections, and for the empirical treatment of persistent neutropenic fever, but not for primary treatment of invasive aspergillosis (IA) or IC.37 

The echinocandin antifungal agents offer yet another alternative to either the azole- or amphotericin B-based regimens.38,39 These cyclic hexapeptide agents inhibit the enzymatic biosynthesis of 1,3-beta glucans in the fungal cell wall of Candida spp., Aspergillus spp., Pneumocystis jiroveci, but not Cryptococcus neoformans or the zygomycetes. A study of caspofungin and amphotericin B deoxycholate in 224 patients with IC suggested a trend favouring caspofungin (73.4% of 109 vs 61.7% of 115, OR 1.71, 95%CI 0.97 to 3.01)40 (Figure 1 ); however, more caspofungin recipients had persistently positive blood cultures with C. parapsilosis (P = 0.02). Another echinocandin, anidulofungin, was found to be superior to fluconazole for IC (75.6% vs 60.2%; difference 15.4%, 95%CI, 3.85 to 26.99, P = 0.01)41(Figure 1 ). A third echinocandin agent, micafungin, is also under study. The safety, efficacy, and availability of these products have diminished the role of polyenes as initial therapy for IC.

Invasive aspergillosis

Primary therapy

The only antifungal agents approved for the treatment of IA are conventional amphotericin B deoxycholate (CAB) and voriconazole, the response rates for the former being of the order of 1 in 3. The seminal study comparing voriconazole to CAB in 277 patients42 demonstrated higher response rates among voriconazole recipients (52.8% versus 31.6%), a 67% improvement (Figure 2 ). Patients with early lesions characterized by pulmonary nodules with halos had higher treatment response rates (52.4% versus 29.1%).43 Moreover, a survival advantage for voriconazole recipients was observed (70.8% compared to 57.9%, a 22% improvement (χ2 = 5.063, P = 0.024)). Despite this, response among allogeneic HSCT recipients remained suboptimal (32.4% for voriconazole versus 13.3% for CAB). A study of dose-intense liposomal amphotericin B (10 mg/kg/d for 14 days followed by 3 mg/kg/d vs 3 mg/kg/d ) as primary therapy for IA demonstrated similar response rates (46% vs 50%, respectively), but more nephrotoxicity (31% versus 14%), hypokalemia (30% versus 16%), and higher mortality in the dose-intense group (41% versus 28%, OR 0.55, 95%CI 0.33 to 0.99).44 Based upon this experience, the value of dose-intensity for IA appears limited.

Salvage monotherapy

The optimal treatment for patients failing or intolerant to primary anti-aspergillus therapy is unknown. Responses in published reports are of the order of 45% (95% CI 40.2% to 50.3%) (Figure 3 ).45,48 

Combination therapy

Poor response rates for primary and salvage monotherapy therapy for IFI and the availability of agents with differing mechanisms of action have fueled recent reviews.49,52 Arguments for considering combination therapy include enhanced fungal killing (synergy), an enhanced spectrum of activity, prevention of development of resistance, and reduction of drug-related toxicities.50 Response rates from retrospective reports in heterogeneous patient populations have been inconsistent.45,53,54 Favorable responses were observed among HSCT patients failing polyene-based therapy for invasive aspergillosis with a combination of voriconazole and caspofungin compared to voriconazole monotherapy.55 Only one recent randomized-controlled trial in IC suggested that a combination of fluconazole and amphotericin B deoxycholate may have advantages over fluconazole monotherapy.34 A preliminary report on a French multicenter randomized study comparing combination therapy with liposomal amphotericin B (3 mg/kg/d) plus caspofungin (70 mg day 1 and 50 mg/d thereafter) versus high-dose liposomal amphotericin B monotherapy (10 mg/kg/d) for primary treatment of invasive aspergillosis was recently presented.56 A favorable overall response was observed in 67% (95%CI 38% to 88%) combination therapy recipients compared to 27% (95%CI 8% to 55%) high-dose therapy recipients (P = 0.028). The results of this small pilot, representing the first prospective study of combination therapy in IA, are encouraging but need confirmation.

Combination antifungal therapies are expensive and potentially toxic and, despite some recent encouraging reports, there are limited well-designed randomized-controlled trials to guide the practicing clinician faced with managing these problems.

Non-Aspergillus mould infections

These have been recently reviewed2,57,58 and include infections due to Fusarium spp., Scedosporium spp., the Zygomycetes, the dematiacious (dark-walled) fungi, Trichoderma spp., Paecilomyces spp., and Scopulariopsis spp. The extremely poor outcomes of infections due to the first three, the most commonly reported pathogens, have been largely a function of selection bias of most profoundly immunosuppressed patients and of reduced antifungal susceptibility profiles to conventional antifungal agents.58 Voriconazole has been effective for infections due to Fusarium spp. and Scedosporium apiospermum.48,59 While concerns have been raised with regard to increased use of voriconazole and risk for Zygomycete infections,59,60 a cause-and-effect relationship remains unproven. Moreover, the rising incidence of Zygomycoses antedated the introduction of this agent into clinical practice.61 Outcomes of posaconazole therapy, an extended-spectrum azole, appear to compare favorably with more standard lipid-based amphotericin B in the treatment of zygomycete infections.62 

Table 1.

Azole-based antifungal prophylaxis in high-risk patients by region and type of agent (fluconazole versus mould-active extended-spectrum azoles).

USA,* 2001Japan,** 2001Europe,† 2005
FLUESAFLUESAFLUESA
*
Trifilio S et al.
Bone Marrow Transplant
.
2004
;
33
:
735
–9
(A survey of 31 transplant centers in the United States encompassing over 3400 transplants performed in 2001. Almost all (93%) reported using some form of antifungal prophylaxis). 
**
Imataki O et al.
Bone Marrow Transplant
.
2004
;
33
:
1173
–9
(A survey among 63 transplant centers encompassing 48% of all HSCT performed in Japan in 2001. All (100%) patients undergoing autologous, myeloablative and nonmyeloablative HSCT received antifungal chemoprophylaxis). 
† Round Table, European Conference on Infections in Leukemia, 14th Symposium on Infections in the Immunocompromised Host, Crans-Montana, Switzerland, July 2–5, 2006. 
Abbreviations: Auto HSCT, autologous hematopoietic stem cell transplant; Allo HSCT, allogeneic hematopoietic stem cell transplant; FLU, fluconazole; ESA, extended-spectrum mould-active azoles (itraconazole, voriconazole, posaconazole). 
Auto HSCT 84% 3% 72% 18% 53% 36% 
Allo HSCT 67% 13% 79% 4% 57% 33% 
Acute leukemia — — — — 55% 35% 
USA,* 2001Japan,** 2001Europe,† 2005
FLUESAFLUESAFLUESA
*
Trifilio S et al.
Bone Marrow Transplant
.
2004
;
33
:
735
–9
(A survey of 31 transplant centers in the United States encompassing over 3400 transplants performed in 2001. Almost all (93%) reported using some form of antifungal prophylaxis). 
**
Imataki O et al.
Bone Marrow Transplant
.
2004
;
33
:
1173
–9
(A survey among 63 transplant centers encompassing 48% of all HSCT performed in Japan in 2001. All (100%) patients undergoing autologous, myeloablative and nonmyeloablative HSCT received antifungal chemoprophylaxis). 
† Round Table, European Conference on Infections in Leukemia, 14th Symposium on Infections in the Immunocompromised Host, Crans-Montana, Switzerland, July 2–5, 2006. 
Abbreviations: Auto HSCT, autologous hematopoietic stem cell transplant; Allo HSCT, allogeneic hematopoietic stem cell transplant; FLU, fluconazole; ESA, extended-spectrum mould-active azoles (itraconazole, voriconazole, posaconazole). 
Auto HSCT 84% 3% 72% 18% 53% 36% 
Allo HSCT 67% 13% 79% 4% 57% 33% 
Acute leukemia — — — — 55% 35% 
Table 2.

Reported initiation and cessation times for antifungal prophylaxis by patient risk group.

Patient GroupStart DateEnd Date
* Day of sustained absolute neutrophil count > 0.5 × 109/L 
Abbreviations: Auto HSCT, autologous hematopoietic stem cell transplant; Allo HSCT, allogeneic hematopoietic stem cell transplant. 
Acute leukemia Day 1 of induction therapy Myeloid reconstitution* 
Auto HSCT Day 1 of conditioning therapy Myeloid engraftment* 
Allo HSCT Day 1 of conditioning therapy
 Transplant day 0
 Transplant day 1 Myeloid engraftment*
 Protocol specified day (+75, +100, +120, +180)
 Cessation of immunosuppressive therapy 
Patient GroupStart DateEnd Date
* Day of sustained absolute neutrophil count > 0.5 × 109/L 
Abbreviations: Auto HSCT, autologous hematopoietic stem cell transplant; Allo HSCT, allogeneic hematopoietic stem cell transplant. 
Acute leukemia Day 1 of induction therapy Myeloid reconstitution* 
Auto HSCT Day 1 of conditioning therapy Myeloid engraftment* 
Allo HSCT Day 1 of conditioning therapy
 Transplant day 0
 Transplant day 1 Myeloid engraftment*
 Protocol specified day (+75, +100, +120, +180)
 Cessation of immunosuppressive therapy 
Figure 1.

Treatment trials of invasive candidiasis. Overall success at end-of-treatment. Also shown are the mean success rates (shaded areas represent the standard deviation of the mean) for trials with amphotericin B deoxycholate and fluconazole arms for comparison.

Figure 1.

Treatment trials of invasive candidiasis. Overall success at end-of-treatment. Also shown are the mean success rates (shaded areas represent the standard deviation of the mean) for trials with amphotericin B deoxycholate and fluconazole arms for comparison.

Close modal
Figure 2.

Antifungal therapy of invasive aspergillosis in myelosuppressed and immunosuppressed cancer patients. Clinical response to primary therapy.

Abbreviations: dAMB, amphotericin B deoxycholate; ABCD, amphotericin B colloidal dispersion; ABLC, amphotericin B lipid complex; L-AMB, liposomal amphotericin B; ITR, itraconazole; VOR, voriconazole.

Figure 2.

Antifungal therapy of invasive aspergillosis in myelosuppressed and immunosuppressed cancer patients. Clinical response to primary therapy.

Abbreviations: dAMB, amphotericin B deoxycholate; ABCD, amphotericin B colloidal dispersion; ABLC, amphotericin B lipid complex; L-AMB, liposomal amphotericin B; ITR, itraconazole; VOR, voriconazole.

Close modal
Figure 3.

Antifungal therapy of invasive aspergillosis in myelosuppressed and immunosuppressed cancer patients. Clinical response to salvage monotherapy.

Abbreviations: ABLC, amphotericin B lipid complex; CAS, caspofungin; ITR, itraconazole; VOR, voriconazole.

Figure 3.

Antifungal therapy of invasive aspergillosis in myelosuppressed and immunosuppressed cancer patients. Clinical response to salvage monotherapy.

Abbreviations: ABLC, amphotericin B lipid complex; CAS, caspofungin; ITR, itraconazole; VOR, voriconazole.

Close modal

Sections of Infectious Diseases and Haematology/Oncology, Professor and Head, Section of Haematology/Oncology, Department of Internal Medicine, The University of Manitoba; Head, Department of Medical Oncology and Haematology, Director, Infection Control Services, CancerCare Manitoba, Winnipeg, Manitoba, Canada

1
Pfaller MA, Pappas PG, Wingard JR. Invasive fungal pathogens: Current epidemiological trends.
Clin Infect Dis
.
2006
;
43
:
S3
–14.
2
Walsh TJ, Groll A, Hiemenz J, et al. Infections due to emerging and uncommon medically important fungal pathogens.
Clin Microbiol Infect
.
2004
;
10
Suppl 1:
48
–66.
3
Alexander BD, Pfaller MA. Contemporary tools for diagnosis and management of invasive mycoses.
Clin Infect Dis
.
2006
;
43
:
S15
–27.
4
Pappas PG, Alexander B, Marr K, et al. Invasive fungal infections (IFIs) in hematopoietic stem cell (HSCTs) and organ transplant recipients (OTRs): Overview of the TRANSNET database [abstract 671]. Boston: IDSA;
2004
:174.
5
Horn D, Fishman J, Anaissie E, et al. PATH Alliance®: A comprehensive, prospective registry of patients with invasive fungal infections [abstract P-024]. In: Anaissie E.J., Rinaldi MG, eds. Vol Presentation Summaries and Abstracts, Focus on Fungal Infections 16 (Las Vegas,March 6–10).
2006
:184.
6
Mennink-Kersten MA, Donnelly JP, Verweij PE. Detection of circulating galactomannan for the diagnosis and management of invasive aspergillosis.
Lancet Infect Dis
.
2004
;
4
:
349
–357.
7
Verweij PE. Advances in diagnostic testing.
Med Mycol
.
2005
;
43
Suppl 1:
S121
–S124.
8
Yang S, Rothman RE. PCR-based diagnostics for infectious diseases: uses, limitations, and future applications in acute-care settings.
Lancet Infect Dis
.
2004
;
4
:
337
–348.
9
Ascioglu S, Rex JH, de Pauw B, et al. Defining opportunistic invasive fungal infections in immunocompromised patients with cancer and hematopoietic stem cell transplants: an international consensus.
Clin Infect Dis
.
2002
;
34
:
7
–14.
10
Subira M, Martino R, Rovira M, et al. Clinical applicability of the new EORTC/MSG classification for invasive pulmonary aspergillosis in patients with hematological malignancies and autopsy-confirmed invasive aspergillosis.
Ann Hematol
.
2003
;
82
:
80
–82.
11
O’Brien SN, Blijlevens NM, Mahfouz TH, Anaissie EJ. Infections in patients with hematological cancer: recent developments.
Hematology (Am Soc Hematol Educ Prog)
.
2003
;
438
–472.
12
Wingard JR, Nichols WG, McDonald GB. Supportive care.
Hematology (Am Soc Hematol Educ Prog)
.
2004
;
372
–389.
13
Bow EJ, Laverdiere M, Lussier N, et al. Antifungal prophylaxis for severely neutropenic chemotherapy recipients: a meta analysis of randomized-controlled clinical trials.
Cancer
.
2002
;
94
:
3230
–3246.
14
Glasmacher A, Prentice A, Gorschluter M, et al. Itraconazole prevents invasive fungal infections in neutropenic patients treated for hematologic malignancies: evidence from a meta-analysis of 3,597 patients.
J Clin Oncol
.
2003
;
21
:
4615
–4626.
15
Cornely O, Maertens J, Winston D, et al. Posaconazole vs standard azole (FLU/ITRA) therapy for prophylaxis of invasive fungal infections (IFIs) among high-risk neutropenic patients: results of a randomized, multicenter trial.
ASH Annual Meeting Abstracts
.
2005
;
106
:
1844
.
16
Ullmann AJ, Lipton JH, Vesole DH, et al. A multicenter trial of oral posaconazole vs. fluconazole for the prophylaxis of invasive fungal infections in recipients of allogeneic hematopoietic stem cell transplantation with graft-vs.-host disease [abstract]. Interscience Conference on Antimicrobial Agents and Chemotherapy
2005
.
17
van Burik JA, Ratanatharathorn V, Stepan DE, et al. Micafungin versus fluconazole for prophylaxis against invasive fungal infections during neutropenia in patients undergoing hematopoietic stem cell transplantation.
Clin Infect Dis
.
2004
;
39
:
1407
–1416.
18
Mattiuzzi GN, Alvarado G, Giles FJ, et al. Open-label, randomized comparison of itraconazole versus caspofungin for prophylaxis in patients with hematologic malignancies.
Antimicrobial Agents Chemother
.
2006
;
50
:
143
–147.
19
Trifilio S, Verma A, Mehta J. Antimicrobial prophylaxis in hematopoietic stem cell transplant recipients: heterogeneity of current clinical practice.
Bone Marrow Transplant
.
2004
;
33
:
735
–739.
20
Imataki O, Kami M, Kim SW, et al. A nationwide survey of deep fungal infections and fungal prophylaxis after hematopoietic stem cell transplantation in Japan.
Bone Marrow Transplant
.
2004
;
33
:
1173
–1179.
21
Marr KA, Crippa F, Leisenring W, et al. Itraconazole versus fluconazole for prevention of fungal infections in patients receiving allogeneic stem cell transplants.
Blood
.
2004
;
103
:
1527
–1533.
22
Yoshida M, Ohno R. Antimicrobial prophylaxis in febrile neutropenia.
Clin Infect Dis
.
2004
;
39
Suppl 1:
S65
–S67.
23
Winston DJ, Maziarz RT, Chandrasekar PH, et al. Intravenous and oral itraconazole versus intravenous and oral fluconazole for long-term antifungal prophylaxis in allogeneic hematopoietic stem-cell transplant recipients. A multicenter, randomized trial.
Ann Intern Med
.
2003
;
138
:
705
–713.
24
Glasmacher A, Prentice AG. Evidence-based review of antifungal prophylaxis in neutropenic patients with haematological malignancies.
J Antimicrob Chemother
.
2005
;
56
Suppl 1:
i23
–i32.
25
Hebart H, Klingspor L, Klingebiehl T, et al. PCR-based liposomal amphotericin B treatment following allogeneic stem cell transplantation is a safe treatment strategy: preliminary results of a prospective study.
ASH Annual Meeting Abstracts
.
2004
;
104
:
192
.
26
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
;
41
:
1242
–1250.
27
Bow EJ, Laverdière M, Rotstein C. A systematic review of the efficacy of azoles and lipid-based formulations of amphotericin B as empirical anti-fungal therapy in persistently febrile neutropenic patients despite broad-spectrum anti-bacterial therapy [abstract].
40th Interscience Conference on Antimicrobial Agents and Chemotherapy
.
2000
;
364
(Abst#702).
28
De Pauw BE. Between over- and undertreatment of invasive fungal disease.
Clin Infect Dis
.
2005
;
41
:
1251
–1253.
29
Kanda Y, Yamamoto R, Chizuka A, et al. Prophylactic action of oral fluconazole against infection in neutropenic patients—a meta-analysis of 16 randomized, controlled trials.
Cancer
.
2000
;
89
:
1611
–1625.
30
Hughes WT, Armstrong D, Bodey GP, et al. 2002 guidelines for the use of antimicrobial agents in neutropenic patients with cancer.
Clin Infect Dis
.
2002
;
34
:
730
–751.
31
Rex JH, Bennett JE, Sugar AM, et al. A randomized trial comparing fluconazole with amphotericin B for the treatment of candidemia in patients without neutropenia. Candidemia Study Group and the National Institute.
N Engl J Med
.
1994
;
331
:
1325
–1330.
32
Anaissie EJ, Darouiche RO, Abi-Said D, et al. Management of invasive candidal infections: results of a prospective, randomized, multicenter study of fluconazole versus amphotericin B and review of the literature.
Clin Infect Dis
.
1996
;
23
:
964
–972.
33
Phillips P, Shafran S, Garber G, et al. Multicenter randomized trial of fluconazole versus amphotericin B for treatment of candidemia in non-neutropenic patients. Canadian Candidemia Study Group.
Eur J Clin Microbiol Infect Dis
.
1997
;
16
:
337
–345.
34
Rex JH, Pappas PG, Karchmer AW, et al. A randomized and blinded multicenter trial of high-dose fluconazole plus placebo versus fluconazole plus amphotericin B as therapy for candidemia and its consequences in nonneutropenic subjects.
Clin Infect Dis
.
2003
;
36
:
1221
–1228.
35
Kontoyiannis DP, Bodey GP, Mantzoros CS. Fluconazole vs. amphotericin B for the management of candidaemia in adults: a meta-analysis.
Mycoses
.
2001
;
44
:
125
–135.
36
Kullberg BJ, Sobel JD, Ruhnke M, et al. Voriconazole versus a regimen of amphotericin B followed by fluconazole for candidaemia in non-neutropenic patients: a randomised non-inferiority trial.
Lancet
.
2005
;
366
:
1435
–1442.
37
Maertens J, Boogaerts M. The place for itraconazole in treatment.
J Antimicrob Chemother
.
2005
;
56
Suppl 1:
i33
–i38.
38
Walsh TJ. Echinocandins—an advance in the primary treatment of invasive candidiasis.
N Engl J Med
.
2002
;
347
:
2070
–2072.
39
Denning DW. Echinocandin antifungal drugs.
Lancet
.
2003
;
362
:
1142
–1151.
40
Mora-Duarte J, Betts R, Rotstein C, et al. Comparison of caspofungin and amphotericin B for invasive candidiasis.
N Engl J Med
.
2002
;
347
:
2020
–2029.
41
Reboli A, Rotstein C, Pappas P, et al. Anidulafungin vs. fluconazole for treatment of candidemia and invasive candidiasis (C/IC) [abstract]. Abstracts of the 45th Interscience Conference on Antimicrobial Agents and Chemotherapy.
2005
.
42
Herbrecht R, Denning DW, Patterson TF, et al. Voriconazole versus amphotericin B for primary therapy of invasive aspergillosis.
N Engl J Med
.
2002
;
347
:
408
–415.
43
Greene R. The radiological spectrum of pulmonary aspergillosis.
Med Mycol
.
2005
;
43
Suppl 1:
S147
–S154.
44
Cornely OA, Maertens J, Bresnik M, Herbrecht R. Liposomal amphotericin B (L-AMB) as initial therapy for invasive filamentous fungal infections (IFFI): a randomized, prospective trial of a high loading regimen vs. standard dosing (AmBiLoad Trial).
ASH Annual Meeting Abstracts
.
2005
;
106
:
3222
.
45
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
–S400.
46
Maertens J, Raad I, Petrikkos G, 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.
47
Caillot D. Intravenous itraconazole followed by oral itraconazole for the treatment of amphotericin-B-refractory invasive pulmonary aspergillosis.
Acta Haematol
.
2003
;
109
:
111
–118.
48
Perfect JR, Marr KA, Walsh TJ, et al. Voriconazole treatment for less-common, emerging, or refractory fungal infections.
Clin Infect Dis
.
2003
;
36
:
1122
–1131.
49
Johnson MD, MacDougall C, Ostrosky-Zeichner L, Perfect JR, Rex JH. Combination antifungal therapy.
Antimicrob Agents Chemother
.
2004
;
48
:
693
–715.
50
Kontoyiannis DP, Lewis RE. Toward more effective antifungal therapy: the prospects of combination therapy.
Br J Haematol
.
2004
;
126
:
165
–175.
51
Mukherjee PK, Sheehan DJ, Hitchcock CA, Ghannoum MA. Combination treatment of invasive fungal infections.
Clin Microbiol Rev
.
2005
;
18
:
163
–194.
52
Cuenca-Estrella M. Combinations of antifungal agents in therapy—what value are they?
J Antimicrob Chemother
.
2004
;
54
:
854
–869.
53
Kontoyiannis DP, Boktour M, Hanna H, et al. Itraconazole added to a lipid formulation of amphotericin B does not improve outcome of primary treatment of invasive aspergillosis.
Cancer
.
2005
;
103
:
2334
–2337.
54
Kontoyiannis DP, Hachem R, Lewis RE, et al. Efficacy and toxicity of caspofungin in combination with liposomal amphotericin B as primary or salvage treatment of invasive aspergillosis in patients with hematologic malignancies.
Cancer
.
2003
;
98
:
292
–299.
55
Marr KA, Boeckh M, Carter RA, Kim HW, Corey L. Combination antifungal therapy for invasive aspergillosis.
Clin Infect Dis
.
2004
;
39
:
797
–802.
56
Caillot D, Thiebault A, Herbrecht R, et al. Liposomal amphotericin B in combination with caspofungin versus liposomal amphotericin B high dose regimen for the treatment of invasive aspergillosis in immunocompromised patients: randmonized pilot study (Combistrat Trial) [abstract]. Focus on Fungal Infections 16 2006;March 8–10,
2006
; Las Vegas, Nevada:174 (Abst P-004).
57
Marr KA, Carter RA, Crippa F, Wald A, Corey L. Epidemiology and outcome of mould infections in hematopoietic stem cell transplant recipients.
Clin Infect Dis
.
2002
;
34
:
909
–917.
58
Nucci M. Emerging moulds: fusarium, scedosporium and zygomycetes in transplant recipients.
Curr Opin Infect Dis
.
2003
;
16
:
607
–612.
59
Nucci M, Marr KA. Emerging fungal diseases.
Clin Infect Dis
.
2005
;
41
:
521
–526.
60
Chamilos G, Marom EM, Lewis RE, Lionakis MS, Kontoyiannis DP. Predictors of pulmonary zygomycosis versus invasive pulmonary aspergillosis in patients with cancer.
Clin Infect Dis
.
2005
;
41
:
60
–66.
61
Kauffman CA. Zygomycosis: reemergence of an old pathogen.
Clin Infect Dis
.
2004
;
39
:
588
–590.
62
Greenberg RN, Mullane K, van Burik JA, et al. Posaconazole as salvage therapy for zygomycosis.
Antimicrob Agents Chemother
.
2006
;
50
:
126
–133.