Abstract
At the 10th European Conference on Infections in Leukaemia (ECIL), the guidelines for antifungal prophylaxis in pediatric and adult patients with hematological malignancies (HM) were updated and some changes introduced. Regarding acute myeloid leukemia (AML) and myelodysplastic syndromes (MDS) patients undergoing remission induction chemotherapy, a B-II grading has been assigned to isavuconazole, micafungin, and caspofungin, based on non-randomized studies that have shown efficacy in preventing invasive fungal diseases (IFD). Regarding high-risk MDS patients treated with azacytidine, prophylaxis with posaconazole during the first four cycles of treatment is supported in the literature. Prophylaxis is not indicated in patients treated for myeloproliferative neoplasms (NPM), acute lymphoid leukemia (ALL), and Hodgkin lymphoma (HL). For patients with chronic lymphocytic leukemia (CLL) and non-Hodgkin lymphoma (NHL), prophylaxis is not generally indicated. For patients with multiple myeloma (MM), prophylaxis is not indicated and the limited epidemiological data available do not support the use of prophylaxis in subjects treated with bispecific antibodies. For patients undergoing allogeneic hematopoietic stem cell transplantation (HSCT), no substantial changes were made, apart from the addition of isavuconazole with grading B-II in the post-engraftment period. In patients undergoing auto-HSCT, antifungal prophylaxis is not indicated. Previous ECIL guidelines did not include CAR-T cells. The expert panel proposes to endorse the use of anti-mold prophylaxis in high-risk patients during pre-infusion and post-infusion, while in low-risk patients, anti-yeast prophylaxis can be recommended (B-II). For pediatric hematology patients, based on newly published data, caspofungin received a B-I grading as mold-active prophylaxis. Moreover, patients with ALL with insufficient treatment response during induction therapy, and children older than 12 y.o are now considered at high risk for IFD and are recommended to receive antifungal prophylaxis.
Similar content being viewed by others
Introduction
In 2005, the European Group for Blood and Marrow Transplantation (EBMT), the European Organization for Research and Treatment of Cancer (EORTC), the European Leukemia Net (ELN), and the International Immunocompromised Host Society (ICHS) inaugurated the European Conference on Infections in Leukemia (ECIL). Its main goal was to establish guidelines or recommendations for the management of infections due to bacteria, viruses, parasites, and fungi in patients with leukemia and in patients undergoing hematopoietic stem cell transplantation (HSCT) [1]. The prevention of invasive fungal disease (IFD) has been one of the key topics from the beginning [1, 2].
The ECIL committee aims to update their guidelines regularly based on current available evidence. During the fifth and sixth meetings (2013 and 2015), guidelines on antifungal prophylaxis for adults were extensively revised, and during the ninth meeting (2021) recommendations for antifungal prophylaxis in pediatrics were developed [1, 3].
An update of previous recommendations was already done in 2018 [4], but over the last few years, several new antineoplastic drugs have been introduced into clinical practice for all hematological malignancies (i.e. BCL-2 inhibitors, FLT-3 inhibitors in acute myeloid leukemias, Bruton Tyrosine Kinase inhibitors (BTKIs) other new tyrosine kinase inhibitors (TKIs), bispecific monoclonal antibodies). In addition, new cellular therapy procedures, such as chimeric-antigen receptor T (CAR-T) cell treatments, are now increasingly used. Therefore, at the ECIL-10 meeting in September 2024, a detailed review of the recent literature was conducted, with the agreed proposals summarized in this review.
Methodology
The methodology of the ECIL conferences has previously been reported [1, 3].
A working group of experts in the field was nominated several months in advance of the biennial conference and was charged with reviewing the literature published since the last update of the guidelines.
A literature review was performed using the PubMed database for publications up from September 2015 and the working group co-authoring this manuscript reviewed all publications identified. Recommendations drawn from data available only as abstracts were provisionally graded, pending the publication of the full papers. The quality of evidence and strength of recommendation were graded according to the EBM grading system of ESCMID (Table 1) [5].
The working group compiled a slide set discussed in several consecutive online group meetings and electronic communication until two weeks before the ECIL-10 plenary meeting held on September 20th, 2024. The final slide set, approved by all group members, was sent by email to all ECIL-10 participants before the plenary. On the day of the meeting (September 20, 2024), the slides were presented by the working group and interactively discussed during a plenary session. The comments made during the plenary discussion were reviewed by the members of the working group in a closed session and recommendations revised accordingly.
The final set of recommendations was thereafter discussed with the ECIL-10 plenary until consensus was reached.
The approved slide set was published on the ECIL website (https://ecil-leukaemia.com/en/resources/resources-ecil), with comments invited for over a month (November 2024). All members of the working group then approved the final set of recommendations.
The final manuscript has been written and revised by all co-authors.
Acute myeloid leukemia (AML)
Two new major issues have been addressed since the publication of the previous ECIL guidelines on antifungal prophylaxis [4].
Oral isavuconazole has been studied for primary antifungal prophylaxis in 65 patients undergoing remission induction chemotherapy in an open-label phase II study [6]. One patient developed a proven IFD and another 3 a probable IFD. When compared with posaconazole and voriconazole in a retrospective analysis of 277 patients with newly diagnosed AML, the incidences of breakthrough IFD were 2.9% for posaconazole, 4.8% for voriconazole, and 5.7% for isavuconazole (p = 0.55) [7]. While isavuconazole is not approved for antifungal prophylaxis in Europe (nor in the United States), the expert panel consider that isavuconazole may be considered for antifungal prophylaxis in selected adult patients undergoing remission induction therapy for AML and for whom posaconazole is not appropriate (e.g., liver function abnormalities, QTc prolongation, drug-drug interactions, intestinal absorption issues).
The benefit from systemic antifungal prophylaxis in patients undergoing consolidation chemotherapy for AML was retrospectively analyzed in a large SEIFEM study [8]. Among 2588 adult and pediatric patients, invasive aspergillosis was diagnosed in 34/1137 (2.9%) patients receiving no antifungal prophylaxis, compared with 22/1451 (1.5%) patients who were given antifungal prophylaxis (p = 0.01). The number needed to treat to prevent one invasive aspergillosis was 71 patients [8]. Systemic antifungal prophylaxis has been given a B-IIu recommendation for AML patient undergoing consolidation chemotherapy.
For patients undergoing AML treatment with one of the newer systemic agents such as venetoclax, FLT3 inhibitors, or ivosidenib, the recommendations on indications and the proper selection of agents for systemic antifungal prophylaxis, as given by the ECIL-9 guideline [9], are reinforced by the present updated guideline. Recommendations for appropriate dose adjustments in case of relevant pharmacological drug-drug interactions were thereby addressed as well.
Beyond these issues, the grading of recommendations has been modified for itraconazole (from B-I to C-I) and micafungin (from C-II to B-II), and a recommendation for “super bioavailable” (SUBA)-itraconazole has been added (C-II).
In comparison to posaconazole, itraconazole was shown to be less effective and less reliable concerning drug levels than posaconazole in AML patients [10, 11]. As SUBA-itraconazole has become available, “classic” itraconazole has been slightly downgraded (C-I).
SUBA-itraconazole has been clinically investigated for pharmacokinetics, tolerability, and safety in several cohort studies including patients with hematologic malignancies and allogeneic HSCT recipients as well as solid organ transplant recipients [12, 13].
In our former recommendations, echinocandins as a group were graded as “C-II”, because of their narrower spectrum of antifungal activity when compared to amphotericin B and most triazoles, and sparse clinical data on their use as antifungal prophylaxis in patients with hematologic malignancies. As more detailed data for the efficacy and safety of micafungin are now available for AML and myelodyplastic syndromes (MDS) patients as well as for allogeneic HSCT recipients, micafungin has now been upgraded to B-II [14,15,16].
Table 2 shows the new recommendations compared to those suggested during ECIL-5.
Myelodysplastic Syndromes (MDS)
For patients with low-risk MDS receiving transfusion-supportive treatment or treated with growth factors (i.e. erythropoietin), no increased risk of IFD has been reported, and therefore no antifungal prophylaxis is indicated (D-I). The introduction of luspatercept does not change this recommendation. The COMMANDS study comparing luspatercept versus erythropoietin which enrolled over 600 patients, did not report any case of IFD [17].
For patients with high-risk MDS receiving intensive AML-like induction (and consolidation) chemotherapy treatment, the recommendation to administer antifungal prophylaxis has not changed (A-I) [4]. The situation is different for intermediate and high-risk patients receiving treatment with azacytidine. A review of the literature in recent years shows an increased risk of IFD, especially during the first 4 cycles of treatment with an incidence ranging from 3% to 12% [18]. When antifungal prophylaxis was administered in most patients, the rate of IFD was 3-8% [18,19,20], while in series where the proportion of patients receiving antifungal prophylaxis was very low, the rate of IFD rised to 8-12% [21,22,23,24,25,26,27]. The new recommendation for these patients is therefore to use antimold prophylaxis during the first 4 cycles of azacytidine treatment (B-IIu).
Chronic myeloproliferative neoplasms
Regarding patients with chronic myeloid leukemia treated with TKIs (imatinib, dasatinib, nilotinib, ponatinib, asciminib) and Philadelphia chromosome-negative myeloproliferative neoplasms, including those treated with ruxolitinib [28,29,30,31,32,33,34] an increased risk of IFD is not reported and antifungal prophylaxis is not indicated (D-I).
Table 3 shows the new recommendations compared to those suggested during ECIL-5.
Acute lymphoblastic leukemia (ALL)
Adults with ALL are usually treated with an intensive polychemotherapy regimen inducing prolonged neutropenia and receive corticosteroids. Therefore, these patients are at risk of developing IFD. The incidence of IFD in this population varies between 4 and 18%, which is rather similar compared to that observed in AML [35]. Although TKIs (e.g. imatinib, nilotinib, ponatinib) used for the treatment of Philadelphia chromosome-positive ALL have been associated with IFD, the actual IFD incidence seems to be low (< 1%) [36]. Novel specific monoclonal antibody therapies used for ALL (e.g. blinatumomab, inotuzumab ozogamicin) have not been associated with a higher IFD incidence compared to the standard of care in randomized controlled trials [37, 38].
Antifungal prophylaxis in this population is hampered by the drug-drug interaction between triazoles and vincristine, an important component of most ALL chemotherapy regimens. Concomitant use of triazoles may result in increased vincristine-related neurotoxicity because this later drug is metabolized by the CYP3A4 cytochrome. Studies in children with ALL suggest a significantly higher risk of vincristine-related neurotoxicity with voriconazole or itraconazole, while the use of fluconazole seems to be safer [39, 40]. However, data on adults are lacking. Studies assessing the efficacy of antifungal prophylaxis in ALL adult patients for the prevention of IFD are heterogeneous and scarce [41,42,43]. One randomized controlled study failed to demonstrate the benefit of twice weekly intravenous liposomal amphotericin B (5 mg/kg) versus placebo with a higher incidence of drug-related adverse events in the treatment arm [44].
Antifungal prophylaxis with a mold-active triazole, such as voriconazole or posaconazole, is not recommended because of interactions which could increase the toxicity of vincristine (D-II). Although data are still lacking, isavuconazole might be considered with caution, considering its lower inhibitory effect on CYP3A4 (C-III). Similarly, fluconazole might be considered with caution for prevention of yeast infection (C-III). Alternative anti-mold prophylaxis (e.g. liposomal amphotericin B, echinocandins) might be considered in high-risk patients (i.e. prolonged chemotherapy-induced neutropenia), but no benefit has been shown to date. No antifungal prophylaxis is recommended for ALL patients receiving only TKIs (D-III) (Table 4).
Chronic lymphocytic leukemia (CLL)
The incidence of IFD in patients with CLL is overall low (0.5 to 3%) and varies according to the type of treatment [45,46,47,48]. These patients present with a large and diverse spectrum of immunosuppression, from untreated patients with different degrees of neutropenia, patients treated with corticosteroids, anti-BCL2 (e.g. venetoclax), or BTKIs (e.g. ibrutinib). Treatment with ibrutinib or other BTKIs is associated with the highest IFI incidence in this population (2 to 3%) [46, 49, 50]. One small retrospective analysis showed a significantly lower IFD incidence among ibrutinib-treated CLL patients receiving antifungal prophylaxis (mainly fluconazole) versus no prophylaxis [51].
Antifungal prophylaxis is not routinely recommended in CLL patients (D-III) but may be considered in selected refractory cases with prolonged neutropenia or BTKIs therapy (C-II). In the case of co-administration of venetoclax, antifungal prophylaxis should be avoided or used cautiously because of drug-drug interactions (i.e. adjustment of venetoclax dosing is needed with therapeutic drug monitoring of the antifungal agent) (Table 4).
Myeloma
Reported IFD incidences in myeloma patients treated with conventional chemotherapy combinations have been low (< 1%) despite the presence of risk factors for IFD such as high doses of corticosteroids, disease-related comorbidities, myeloma-related innate immunodeficiency and, in treatment-experienced patients, poor marrow function [52]. Since the publication of the previous ECIL guidelines, treatment with combinations of immunomodulatory drugs, proteasome inhibitors, monoclonal antibodies, and autologous HSCT have been the standard of care. While no prospective study specifically reporting on IFD in this setting has been published, retrospective studies have reported a somewhat higher IFD incidence than with conventional chemotherapy, 2.7%, 3.5%, and 5.6%, respectively [46, 53,54,55,56,57,58,59,60]. Recently, two new types of antibody treatments have been introduced, bispecific antibodies activating a T-cell response by binding to both myeloma cells and T-cells, and B-cell binding antibodies conjugated with a cytotoxic compound (antibody-drug compound, ADC). Although neither prospective trials nor retrospective studies of treatment with bispecific antibodies have reported the exact numbers of IFD, their overall incidence after excluding Pneumocystis pneumonia has been low (< 2%) [47, 61,62,63,64]. The only registered ADC is belantamab mafotidin, which has now been withdrawn as a single agent but is currently under consideration as part of a combination treatment. The incidence of IFD was not specifically reported in the treatment trials leading to its registration, but the total infection rates were low with a 3% total incidence of pulmonary infections in the largest trial including 218 patients [61]. Routine antifungal prophylaxis is thus not recommended in myeloma patients, regardless of treatment with bispecific antibodies (D-II) (Table 4). Expert panels suggest to consider mold active prophylaxis in high-risk populations such as prolonged neutropenia or prolonged steroid treatment or secondary prophylaxis (no trials).
Non-hodgkin lymphoma (NHL)
Patients with NHL have an overall low IFD incidence (0.5 to 3%) [46, 47]. The incidence among NHL patients receiving BKTIs (e.g. ibrutinib) is roughly similar (about 1.5%) [49]. Some factors have been associated with a higher risk of IFD, such as primary refractoriness, use of two or more previous treatment lines, and occurrence of neutropenia [62].
Antifungal prophylaxis is not routinely recommended in patients with NHL (D-II) but might be considered in selected patients with refractory lymphoma and/or repeated intensive chemotherapies with neutropenia or high dose steroids or BTKI therapy (C-II) (Table 4).
Hodgkin lymphoma (HL)
The risk for IFD tends to be low in patients with Hodgkin lymphoma. Two recent nationwide epidemiological studies in hospitalized HL patients reported a total IFD incidence of 0.5% in Australia and a 0.5% incidence of pulmonary aspergillosis in Spain [47, 63]. In line with previous recommendations, routine antifungal prophylaxis is not recommended (D-II) (Table 4).
Allogeneic HSCT (allo-HSCT)
The main practice change since the previous ECIL recommendations [4] has been the development of haplo-identical allo-HSCT using post-transplantation cyclophosphamide (haplo/PTCy) as graft-versus-host-disease (GVHD) prophylaxis. Retrospective studies on haplo/PTCy allo-HSCT report a one-year incidence rate of IFD ranging from 6 to 17% [64,65,66,67,68]. In two retrospective studies, the IFD incidence (especially invasive mold infections) was significantly higher in haplo/PTCy than in patients transplanted from HLA-matched related and/or unrelated donors receiving calcineurin inhibitors with or without anti-thymocyte globulin (ATG) [66, 67]. However, as the reported IFD rates remained within the range of those in post allo-HSCT outside of the haplo/PTCy setting, haplo/PTCy was still considered at low risk of IFD by the expert panel. As previously highlighted, [4] allo-HSCT centers should monitor the incidence and epidemiology of IFD and be aware that construction works may alter environmental exposure, which may warrant local adaptation of primary antifungal prophylaxis strategy.
The use of isavuconazole as primary antifungal prophylaxis in allo-HSCT recipients has been reported in two prospective open-label studies [69, 70]. The reported rates of breakthrough IFD were low (3–5%) while the treatment was well tolerated (discontinuation rate for toxicity: 2–7%). Data are insufficient to recommend isavuconazole as first-line prophylaxis; however, the expert panel proposes to endorse the ASTCT (American Society of Transplantation and Cellular Therapy) recommendations allowing the use of isavuconazole in cases of QT prolongation, or intolerance to voriconazole or posaconazole (B-II) [71].
In Tables 5 and 6, the main changes in antifungal prophylaxis recommendation are reported (with two additional references in Table 5 [72, 73]).
Anti-CD19 chimeric antigen receptor (CAR) T cells
Previously published ECIL guidelines did not include CAR-T cells [4]. The EBMT/JACIE (Joint Acreditation Committee International Society for Cellular Therapy (ISTC) EBMT) and EBMT/ASTCT guidelines recommend the use of an anti-Candida prophylaxis and suggest discussing an anti-mold prophylaxis in case of prolonged neutropenia and/or steroid use [74, 75].
Retrospective studies, along with two literature reviews, report a 1–15% incidence of IFD in patients treated with anti-CD19 CAR-T cells [76,77,78,79,80]. These studies highlight a significant association between the occurrence of cytokine release syndrome (CRS) and a higher incidence of infections, attributed to the use of systemic immunosuppressive agents. In the literature review by Garner et al. [80], a combination of pre-and post-infusion factors seemed to increase the risk of IFD. The panel endorses the proposal of Garner et al., published after the EBMT/ASTCT recommendations, and integrating new data not available at the time of publication of the EBMT/ASTCT recommendations. The use of anti-mold prophylaxis is thus proposed to patients with pre-infusion (such as neutropenia, previous IFD, previous allo-HSCT, refractory disease) and post-infusion (CRS/immune effector cell-associated neurotoxicity syndrome [ICANS] necessitating steroid therapy and/or tocilizumab, prolonged neutropenia, use of alternative immunosuppressive agent) risk factors for mold infections, while patients without these risk factors could receiveanti-yeast prophylaxis (B-II) [80].
Autologous HSCT
In the previous ECIL recommendations, patients undergoing autologous HSCT, for whatever underlying condition, were considered at low risk of IFD. Primary antifungal prophylaxis is not recommended, although fluconazole (400 mg q24h) should be considered to prevent mucosal Candida infection during the neutropenic phase (B-III) [4]. There are no recommendations for change for autologous HSCT recipients.
Primary antifungal prophylaxis in pediatric hematological malignancies
Mold-active antifungal prophylaxis is recommended for pediatric patients at high risk of IFD (incidence ≥10%), encompassing a subgroup of children with ALL, though their specific risk profile is less precisely defined [81].
A recent analysis of 6316 children with ALL enrolled in the international AIEOP-BFM ALL2009 trial reported an incidence of proven/probable IFDs at 3.8%, with a 12-week mortality rate of 11.2% [82]. In this cohort, 68% of infections were mold-related, with significant risk factors for IFD being ≥12 years of age and insufficient treatment response. A diagnoses of proven/probable IFD were associated with a elevated hazard ratio for event-free survival and overall survival [82]. Consequently, older children (≥ 12 years) with ALL and those with insufficient treatment response are identified as being at elevated risk for IFD and are now recommended to receive antifungal prophylaxis.
Since the 2021 update by ECIL, advances in pediatric antifungal development have been significant. A recent multicenter, non-randomized, open-label, phase 1b dose-escalation trial demonstrated that posaconazole intravenous solution (IV) and powder for oral suspension (PFS) were well tolerated in children, with safety profiles similar to adults [83]. Following this dose-finding trial, the European Medicines Agency (EMA) approved posaconazole IV and PFS in 2021 for high-risk patients aged ≥2 years and weighing ≤40 kg, including those with AML/MDS or undergoing HSCT with GVHD [84]. Delayed-release tablets were approved for patients ≥2 years and >40 kg with the same conditions [84], although oral suspension remains unapproved by EMA [85].
Additionally, a prospective, randomized, open-label trial among pediatric allo-HSCT patients found caspofungin to be as effective as voriconazole and other triazoles in preventing IFD, including aspergillosis, with a 1.4% infection rate across both trial arms [86]. Hence, the caspofungin grade of recommendation has been updated to a B-I.
Future prospects
The characteristics of an ‘ideal’ agent for antifungal prophylactic use include (a) broad spectrum of activity - covering both yeast and mold pathogens - with low risk of development of resistance, (b) availability in oral and parenteral formulations, (c) low potential for clinically problematic drug-drug interactions, (d) low risk of acute and chronic treatment-limiting toxicities, and (e) predictable pharmacokinetics. Several molecules with antifungal activity with a novel mechanism of action are currently in various stages of clinical development [87]. These new molecules tackle some of the shortcomings of the currently available aramamentarium.
-
Rezafungin, a second generation echinocandin with enhanced PK/PD pharmacometrics, is active in vitro against most wild-type and azole-resistant Candida species (including C. auris), Aspergillus species (including azole-resistant A. fumigatus and cryptic species) and Pneumocystis jirovecii. The drug has minimal risk of drug-drug interactions and has recently been approved for the treatment of candidemia and invasive candidiasis [88]. Its prophylactic efficacy and safety, when given intravenously once weekly, is currently being tested in a phase 3 randomized double-blind study versus a standard antimicrobial regimen (including fluconazole/posaconazole plus trimethoprim-sulfamethoxazole) in allo-HSCT recipients (The ReSPECT Study) [89].
-
Ibrexafungerp, a first-in-class oral glucan synthase inhibitor (a triterpenoid), is approved for the treatment of recurrent vulvovaginal Candida infection. The spectrum of activity is similar to the spectrum of rezafungin, but also includes Alternaria and Cladosporium species. The drug is generally well tolerated with a low risk for drug-drug interactions [90]. Ibrexafungerp has not yet been studied as prophylactic agent but has the potential for use in primary prophylaxis (similar to rezafungin).
-
The fungicidal orotomide olorofim is a potent inhibitor of fungal dihydroorotate dehydrogenase. The drug is given orally, has a good tissue distribution and is generally well tolerated. Although olorofim has activity against a variety of mold pathogens (excluding Mucorales species) and dimorphic fungi, the drug displays no activity against yeast pathogens [91]. As such, olorofim is not a good candidate for primary prophylaxis, but may be used for secondary prophylaxis in patients with well documented prior mold infections (e.g., scedosporiosis or aspergillosis).
-
Fosmanogepix (the active moiety is manogepix) targets fungal glycosylphosphatidylinositol-anchored cell wall transfer protein 1, inhibiting cell wall synthesis causing loss of viability. Manogepix has a very broad spectrum of activity covering most clinically important fungal pathogens. The drug is available as an oral and IV formulation, has a wide tissue distribution and displays linear pharmacokinetics. The drug has favorable drug-drug interaction and safety profiles [92]. A phase 1 safety and PK study has been performed in neutropenic leukemia patients receiving the drug prophylactically [93]. Given these characteristics, the drug has potential for being investigated as prophylactic antifungal agent.
-
Opelconazole is an azole with activity against Aspergillus species and other fungi including various Candida sp. (including C. auris), Rhizopus oryzae, Cryptococccus neoformans, Chaetomium globosum, Penicillium chrysogenum and Trichophyton rubrum. Opelconazole was specifically designed for inhaled delivery; the drug accumulates in the lung and has a long residence time in airway cells, potentially enhancing the ability of host cells to clear the fungus, both in treatment and in prophylaxis. Systemic exposure is very low (ratio of lung: systemic concentrations is ~7000:1), resulting in a low risk for drug-drug interactions [94].
Conclusion
IFDs remain potentially fatal events in patients with hematological malignancies undergoing chemotherapy, transplantation or cellular therapies. Identification of the main risk factors is necessary to establish appropriate antifungal prophylaxis.
In recent years, various antineoplastic drugs directed against specific surface proteins or molecular targets have been developed for almost all hematological malignancies. This has opened new therapeutic possibilities, but has also increased the population at risk of developing IFDs. Development of cellular therapies, i.e. CAR-T therapy, and increasing use of allo-HSCT has further expanded the number of patients at risk of IFD. The main differences in this updated ECIL guidelines is the inclusion of recommendations for these new risk groups, such as venetoclax combined with azacytidine in patients with AML, treatment with bispecific antibiodies, and CAR-T cell therapies.
We aimed to give solid indications based on randomized clinical trials, but in some cases, in the absence of randomized trials, clinical evidence from observational studies has been the base for a grading of recommendation (i.e. isavuconazole in AML).
New bispecific monoclonal antibodies have been introduced during the past 5 years for the treatment of myelomas and lymphomas, but current knowledge on the possible epidemiology of IFDs in these settings is still very limited and does not allow us to suggest any recommendation.
Until now, in Europe, lymphoma patients have been identified as receiving the most benefit from CAR-T cell therapies. However, the number of these procedures is constantly increasing, including myeloma and ALL patients, while their timing is changing, shifting from “the last line therapeutic change” to earlier treatment settings, and now being also used as a bridge to allo-HSCT.
The expectations for the near future are the introduction of new antineoplastic agents that will be more effective but which will also lead to increased immunosuppression. At the same time, we expect that new antifungals with greater efficacy and fewer pharmacological interactions will be available, which will hopefully have an important impact not only on the treatment of IFD but also on prophylaxis.
References
Cordonnier C, Calandra T. The first European Conference on Infections in Leukaemia: why and how?. Eur J Cancer. 2007;5:2–4–4.
Maertens JA, Frère P, Lass-Flörl C, Heinz W, Cornely OA. Primary antifungal prophylaxis in leukaemia patients. Eur J Cancer. 2007;5:43–48–8.
Maertens J, Marchetti O, Herbrecht R, Cornely OA, Flückiger U, Frêre P, et al. European guidelines for antifungal management in leukemia and hematopoietic stem cell transplant recipients: summary of the ECIL 3–2009 update. Bone Marrow Transpl. 2011;46:709–18.
Maertens JA, Girmenia C, Brüggemann RJ, Duarte RF, Kibbler CC, Ljungman P, et al. European guidelines for primary antifungal prophylaxis in adult haematology patients: summary of the updated recommendations from European Conference on Infections in Leukemia. J Antimicrob Chemother. 2018;73:3221–30.
Ullmann AJ, Akova M, Herbrecht R, Viscoli C, Arendrup MC, Arikan-Akdagli S, et al. ESCMID* guideline for the diagnosis and management of Candida diseases 2012: adults with haematological malignancies and after haematopoietic stem cell transplantation (HCT). Clin Microbiol Infect. 2012;18:53–67.
Bose P, McCue D, Wurster S, Wiederhold NP, Konopleva M, Kadia TM, et al. Isavuconazole as primary antifungal prophylaxis in patients with acute myeloid leukemia or myelodysplastic syndrome: an open-label, prospective, phase 2 study. Clin Infect Dis. 2021;72:1755–63.
Rausch CR, DiPippo AJ, Jiang Y, DiNardo CD, Kadia T, Maiti A, et al. Comparison of mold active triazoles as primary antifungal prophylaxis in patients with newly diagnosed acute myeloid leukemia in the era of molecularly targeted therapies. Clin Infect Dis. 2022;75:1503–10.
Del Principe MI, Dragonetti G, Verga L, Candoni A, Marchesi F, Cattaneo C, et al. ‘Real-life’ analysis of the role of antifungal prophylaxis in preventing invasive aspergillosis in AML patients undergoing consolidation therapy: Sorveglianza Epidemiologica Infezioni nelle Emopatie (SEIFEM) 2016 study. J Antimicrob Chemother. 2019;74:1062–8.
Maschmeyer G, Bullinger L, Garcia-Vidal C, Herbrecht R, Maertens J, Menna P, et al. Infectious complications of targeted drugs and biotherapies in acute leukemia. Clinical practice guidelines by the European Conference on Infections in Leukemia (ECIL), a joint venture of the European Group for Blood and Marrow Transplantation (EBMT), the European Organization for Research and Treatment of Cancer (EORTC), the International Immunocompromised Host Society (ICHS) and the European Leukemia Net (ELN). Leukemia. 2022;36:1215–26.
Kallee S, Scharf C, Schroeder I, Paal M, Vogeser M, Irlbeck M, et al. Comparing posaconazole and itraconazole for antifungal prophylaxis in critically ill lung transplant recipients: Efficacy and plasma concentrations. Transpl Infect Dis. 2021;23:e13675.
Copley MS, Waldron M, Athans V, Welch SC, Brizendine KD, Cober E, et al. Itraconazole vs. posaconazole for antifungal prophylaxis in patients with acute myeloid leukemia undergoing intensive chemotherapy: A retrospective study. Int J Antimicrob Agents. 2020;55:105886.
Nield B, Larsen SR, van Hal SJ. Clinical experience with new formulation SUBA®-itraconazole for prophylaxis in patients undergoing stem cell transplantation or treatment for haematological malignancies. J Antimicrob Chemother. 2019;74:3049–55.
Whitmore TJ, Yaw M, Lavender M, Musk M, Boan P, Wrobel J. A novel highly bio-available itraconazole formulation (SUBA®-Itraconazole) for anti-fungal prophylaxis in lung transplant recipients. Transpl Infect Dis. 2021;23:e13587.
Epstein DJ, Seo SK, Huang YT, Park JH, Klimek VM, Berman E, et al. Micafungin versus posaconazole prophylaxis in acute leukemia or myelodysplastic syndrome: A randomized study. J Infect. 2018;77:227–34.
Wingen-Heimann SM, Cornely OA, Vehreschild MJGT, Wisplinghoff H, Franke B, Schons M, et al. Clinical and pharmacoeconomic evaluation of antifungal prophylaxis with continuous micafungin in patients undergoing allogeneic stem cell transplantation: A six-year cohort analysis. Mycoses. 2021;64:437–44.
Kang WH, Song GW, Lee SG, Suh KS, Lee KW, Yi NJ, et al. A multicenter, randomized, open-label study to compare micafungin with fluconazole in the prophylaxis of invasive fungal infections in living-donor liver transplant recipients. J Gastrointest Surg. 2020;24:832–40.
Platzbecker U, Della Porta MG, Santini V, Zeidan AM, Komrokji RS, Shortt J, et al. Efficacy and safety of luspatercept versus epoetin alfa in erythropoiesis-stimulating agent-naive, transfusion-dependent, lower-risk myelodysplastic syndromes (COMMANDS): interim analysis of a phase 3, open-label, randomised controlled trial. Lancet. 2023;402:327–85.
Lorenzana N, Avila LF, Alonso S, Colado E, Bernal T. The impact of antimicrobial prophylaxis in morbidity and infections during azacitidine treatment. Ann Hematol. 2017;96:1833–40.
Tey A, Shaw B, Cardamone L, Shepherd S, Paul E, Rogers B, et al. Risk factors for invasive fungal infection in 5-azacytidine treated patients with acute myeloid leukemia and myelodysplastic syndrome. Eur J Haematol. 2021;107:181–9.
Kang KW, Lee BH, Jeon MJ, Yu ES, Sik Kim D, Lee SR, et al. Efficacy of posaconazole prophylaxis in acute myeloid leukemia and myelodysplastic syndrome patients treated with hypomethylating agents. Ther Adv Hematol. 2020;11:2040620720966882.
Pomares H, Arnan M, Sánchez-Ortega I, Sureda A, Duarte RF. Invasive fungal infections in AML/MDS patients treated with azacitidine: a risk worth considering antifungal prophylaxis?. Mycoses. 2016;59:516–9.
Schuck A, Goette MC, Neukirchen J, Kuendgen A, Gattermann N, Schroeder T, et al. A retrospective study evaluating the impact of infectious complications during azacitidine treatment. Ann Hematol. 2017;96:1097–104.
Trubiano JA, Dickinson M, Thursky KA, Spelman T, Seymour JF, Slavin MA, et al. Incidence, etiology and timing of infections following azacitidine therapy for myelodysplastic syndromes. Leuk Lymphoma. 2017;58:2379–86.
Mądry K, Lis K, Biecek P, Młynarczyk M, Rytel J, Górka M, et al. Predictive model for infection risk in myelodysplastic syndromes, acute myeloid leukemia, and chronic myelomonocytic leukemia patients treated with azacitidine; azacitidine infection risk model: the polish adult leukemia group study. Clin Lymphoma Myeloma Leuk. 2019;19:264–74.e4.
Latagliata R, Niscola P, Fianchi L, Aloe Spiriti MA, Maurillo L, Carmosino I, et al. Pulmonary infections in patients with myelodysplastic syndromes receiving frontline azacytidine treatment. Hematol Oncol. 2020;38:189–96.
Kim GYG, Burns J, Freyer CW, Hamilton KW, Frey NV, Gill SI, et al. Risk of invasive fungal infections in patients with high-risk MDS and AML receiving hypomethylating agents. Am J Hematol. 2020;95:792–8.
Kirkizlar TA, Kirkizlar O, Demirci U, Umut A, Iflazoglu H, Umit EG, et al. Incidence and predisposing factors of infection in patients treated with hypomethylating agents. Leuk Res. 2023;127:107043.
Polverelli N, Breccia M, Benevolo G, Martino B, Tieghi A, Latagliata R, et al. Risk factors for infections in myelofibrosis: role of disease status and treatment. A multicenter study of 507 patients. Am J Hematol. 2017;92:37–41.
Landtblom AR, Andersson TM, Dickman PW, Smedby KE, Eloranta S, Batyrbekova N, et al. Risk of infections in patients with myeloproliferative neoplasms-a population-based cohort study of 8363 patients. Leukemia. 2021;35:476–84.
Breccia M, Andriani A, Montanaro M, Abruzzese E, Buccisano F, Cedrone M, et al. Ruxolitinib in clinical practice for primary and secondary myelofibrosis: an analysis of safety and efficacy of Gruppo Laziale of Ph-negative MPN. Ann Hematol. 2017;96:387–91.
Palandri F, Tiribelli M, Benevolo G, Tieghi A, Cavazzini F, Breccia M, et al. Efficacy and safety of ruxolitinib in intermediate-1 IPSS risk myelofibrosis patients: Results from an independent study. Hematol Oncol. 2018;36:285–90.
Polverelli N, Palumbo GA, Binotto G, Abruzzese E, Benevolo G, Bergamaschi M, et al. Epidemiology, outcome, and risk factors for infectious complications in myelofibrosis patients receiving ruxolitinib: A multicenter study on 446 patients. Hematol Oncol. 2018. https://doi.org/10.1002/hon.2509.
Sylvine P, Thomas S, Pirayeh E. French network of regional pharmacovigilance centers. infections associated with ruxolitinib: study in the french pharmacovigilance database. Ann Hematol. 2018;97:913–4.
Tremblay D, King A, Li L, Moshier E, Coltoff A, Koshy A, et al. Risk factors for infections and secondary malignancies in patients with a myeloproliferative neoplasm treated with ruxolitinib: a dual-center, propensity score-matched analysis. Leuk Lymphoma. 2020;61:660–7.
Stafylidis C, Diamantopoulos P, Athanasoula E, Solomou E, Anastasopoulou A. 2022. Acute lymphoblastic leukemia and invasive mold infections: a challenging field. J Fungi. 2022;8:1127.
Gold JAW, Tolu SS, Chiller T, Benedict K, Jackson BR. Incidence of invasive fungal infections in patients initiating ibrutinib and other small molecule kinase Inhibitors-United States, July 2016-June 2019. Clin Infect Dis. 2022;75:334–7.
Kantarjian H, Stein A, Gökbuget N, Fielding AK, Schuh AC, Ribera JM, et al. Blinatumomab versus chemotherapy for advanced acute lymphoblastic leukemia. N Engl J Med. 2017;376:836–47.
Kantarjian HM, DeAngelo DJ, Stelljes M, Liedtke M, Stock W, Gökbuget N, et al. Inotuzumab ozogamicin versus standard of care in relapsed or refractory acute lymphoblastic leukemia: Final report and long-term survival follow-up from the randomized, phase 3 INO-VATE study. Cancer. 2019;125:2474–87.
van Schie RM, Bruggemann RJ, Hoogerbrugge PM, te Loo DM. Effect of azole antifungal therapy on vincristine toxicity in childhood acute lymphoblastic leukaemia. J Antimicrob Chemother. 2011;66:1853–6.
Yang L, Yu L, Chen X, Hu Y, Wang B. Clinical analysis of adverse drug reactions between vincristine and triazoles in children with acute lymphoblastic leukemia. Med Sci Monit. 2015;21:1656–61.
Doan TN, Kirkpatrick CM, Walker P, Slavin MA, Ananda-Rajah MR, Morrissey CO, et al. Primary antifungal prophylaxis in adult patients with acute lymphoblastic leukaemia: a multicentre audit. J Antimicrob Chemother. 2016;71:497–505.
Oh SM, Byun JM, Chang E, Kang CK, Shin DY, Koh Y, et al. Incidence of invasive fungal infection in acute lymphoblastic and acute myelogenous leukemia in the era of antimold prophylaxis. Sci Rep. 2021;11:22160.
Wang Y, Xing Y, Chen L, Meng T, Li Y, Xie J, et al. Fluconazole versus mould-active triazoles for primary antifungal prophylaxis in adult patients with acute lymphoblastic leukemia: clinical outcome and cost-effectiveness analysis. Int J Hematol. 2018;107:235–43.
Cornely OA, Leguay T, Maertens J, Vehreschild M, Anagnostopoulos A, Castagnola C, et al. Randomized comparison of liposomal amphotericin B versus placebo to prevent invasive mycoses in acute lymphoblastic leukaemia. J Antimicrob Chemother. 2017;72:2359–67.
Infante SM, Fernandez-Cruz A, Nunez L. Severe infections in patients with lymphoproliferative diseases treated with new targeted drugs: A multicentric real-world study. Cancer Med. 2021;10:7629–40.
Tisi MC, Hohaus S, Cuccaro A, Innocenti I, De Carolis E, Za T, et al. Invasive fungal infections in chronic lymphoproliferative disorders: a monocentric retrospective study. Haematologica. 2017;102:e108–e11.
Valentine JC, Morrissey CO, Tacey MA, Liew D, Patil S, Peleg AY, et al. A population-based analysis of invasive fungal disease in haematology-oncology patients using data linkage of state-wide registries and administrative databases: 2005-2016. BMC Infect Dis. 2019;19:274.
Visentin A, Gurrieri C, Imbergamo S, Lessi F, Di Maggio SA, Frezzato F, et al. Epidemiology and risk factors of invasive fungal infections in a large cohort of patients with chronic lymphocytic leukemia. Hematol Oncol. 2017;35:925–8.
Allouchery M, Brunet K, Tomowiak C, Singier A, Pambrun E, Pariente A, et al. Invasive fungal infection incidence and risk factors in patients receiving ibrutinib in real-life settings: A nationwide population-based cohort study. Mycoses. 2024;67:e13676.
Holowka T, Cheung H, Malinis M, Gan G, Deng Y, Perreault S, et al. Incidence and associated risk factors for invasive fungal infections and other serious infections in patients on ibrutinib. J Infect Chemother. 2021;27:1700–5.
Yasu T, Sakurai K, Hoshino M, Akazawa M. Oral azole antifungal prophylaxis in Japanese patients with chronic lymphocytic leukemia receiving ibrutinib: a nationwide cohort study. Leuk Lymphoma. 2023;64:730–3.
Pagano L, Caira M, Candoni A, Offidani M, Fianchi L, Martino B, et al. The epidemiology of fungal infections in patients with hematologic malignancies: the SEIFEM-2004 study. Haematologica. 2006;91:1068–75.
Lim C, Sinha P, Harrison SJ, Quach H, Slavin MA, Teh BW. Low rates of invasive fungal disease in patients with multiple myeloma managed with new generation therapies: Results from a multi-centre cohort study. Mycoses. 2021;64:30–4.
Tsai CK, Liu YC, Kuan AS, Lee KL, Yeh CM, Lee YT, et al. Risk and impact of invasive fungal infections in patients with multiple myeloma. Ann Hematol. 2020;99:1813–22.
Bahlis NJ, Costello CL, Raje NS, Levy MY, Dholaria B, Solh M, et al. Elranatamab in relapsed or refractory multiple myeloma: the MagnetisMM-1 phase 1 trial. Nat Med. 2023;29:2570–6.
Chari A, Minnema MC, Berdeja JG, Oriol A, van de Donk N, Rodríguez-Otero P, et al. Talquetamab, a T-Cell-Redirecting GPRC5D Bispecific Antibody for Multiple Myeloma. N Engl J Med. 2022;387:2232–44.
Lesokhin AM, Tomasson MH, Arnulf B, Bahlis NJ, Miles Prince H, Niesvizky R, et al. Elranatamab in relapsed or refractory multiple myeloma: phase 2 MagnetisMM-3 trial results. Nat Med. 2023;29:2259–67.
Mohan M, Monge J, Shah N, Luan D, Forsberg M, Bhatlapenumarthi V, et al. Teclistamab in relapsed refractory multiple myeloma: multi-institutional real-world study. Blood Cancer J. 2024;14:35.
Nooka AK, Rodriguez C, Mateos MV, Manier S, Chastain K, Banerjee A, et al. Incidence, timing, and management of infections in patients receiving teclistamab for the treatment of relapsed/refractory multiple myeloma in the MajesTEC-1 study. Cancer. 2024;130:886–900.
Uttervall K, Tätting L, Lemonakis K, Majd M, Crafoord J, Olsson M, et al. Effectiveness and infectious complications of BCMA T-cell engagers in treating multiple myeloma: Real-world evidence from Sweden. Cancer Med. 2024;13:e7048.
Dimopoulos MA, Hungria VTM, Radinoff A, Delimpasi S, Mikala G, Masszi T, et al. Efficacy and safety of single-agent belantamab mafodotin versus pomalidomide plus low-dose dexamethasone in patients with relapsed or refractory multiple myeloma (DREAMM-3): a phase 3, open-label, randomised study. Lancet Haematol. 2023;10:e801–12.
Takaoka K, Nannya Y, Shinohara A, Arai S, Nakamura F, Kurokawa M. A novel scoring system to predict the incidence of invasive fungal disease in salvage chemotherapies for malignant lymphoma. Ann Hematol. 2014;93:1637–44.
Tamayo-Velasco Á, López-Herrero R, Gómez-García LM, Sánchez-de Prada L, Aguilar-Monserrate G, Martín-Fernández M, et al. COVID-19-associated pulmonary aspergillosis (CAPA) in hematological patients: Could antifungal prophylaxis be necessary? A nationwide study. J Infect Public Health. 2024;17:939–46.
Fayard A, Daguenet E, Blaise D, Chevallier P, Labussière H, Berceanu A, et al. Evaluation of infectious complications after haploidentical hematopoietic stem cell transplantation with post-transplant cyclophosphamide following reduced-intensity and myeloablative conditioning: a study on behalf of the francophone society of stem cell transplantation and cellular therapy (SFGM-TC). Bone Marrow Transpl. 2019;54:1586–94.
Lien MY, Yeh SP, Gau JP, Wang PN, Li SS, Dai MS, et al. High rate of invasive fungal infections after non-t cell depleted haploidentical allo-HSCT even under antifungal prophylaxis. Bone Marrow Transpl. 2021;56:1750–53.
Busca A, Cinatti N, Gill J, Passera R, Dellacasa CM, Giaccone L, et al. Management of invasive fungal infections in patients undergoing allogeneic hematopoietic stem cell transplantation: the turin experience. Frontiers in Cellular and Infection. Microbiology. 2021;11:805514.
Papanicolaou GA, Chen M, He N, Martens MJ, Kim S, Batista MV, et al. ‘Incidence and impact of fungal infections in post-transplantation cyclophosphamide-based graft-versus-host disease prophylaxis and haploidentical hematopoietic cell transplantation: a center for international blood and marrow transplant research analysis’. Transpl Cell Ther. 2024;30:114.e1–114.e16.
Little JS, Shapiro RM, Aleissa MM, Kim A, Chang J, Kubiak DW, et al. ‘Invasive yeast infection after haploidentical donor hematopoietic cell transplantation associated with cytokine release syndrome’. Transpl Cell Ther. 2022;28:508.e1–508.e8.
Stern A, Su Y, Lee YJ, Seo S, Shaffer B, Tamari R, et al. A Single-center, open-label trial of isavuconazole prophylaxis against invasive fungal infection in patients undergoing allogeneic hematopoietic cell transplantation. Biol Blood Marrow Transpl. 2020;26:1195–202.
Nguyen MH, Ostrosky-Zeichner L, Pappas PG, Walsh TJ, Bubalo J, Alexander BD, et al. Real-World Use of Mold-Active Triazole Prophylaxis in the Prevention of Invasive Fungal Diseases: Results from a Subgroup Analysis of a Multicenter National Registry. Open Forum Infect Dis. 2020;10:ofad424.
Dadwal SS, Hohl TM, Fisher CE, Boeckh M, Papanicolaou G, Carpenter PA, et al. American Society of Transplantation and Cellular Therapy Series, 2: Management and Prevention of Aspergillosis in Hematopoietic Cell Transplantation Recipients. Transpl Cell Ther. 2021;27:201–11.
Girmenia C, Raiola AM, Piciocchi A, Algarotti A, Stanzani M, Cudillo L, et al. Incidence and outcome of invasive fungal diseases after allogeneic stem cell transplantation: a prospective study of the Gruppo Italiano Trapianto Midollo Osseo (GITMO). Biol Blood Marrow Transpl. 2014;20:872–80.
Puerta-Alcalde P, Champlin RE, Kontoyiannis DP. How I perform hematopoietic stem cell transplantation on patients with a history of invasive fungal disease. Blood. 2020;136:2741–53.
Yakoub-Agha I, Chabannon C, Bader P, Basak GW, Bonig H, Ciceri F, et al. Management of adults and children undergoing chimeric antigen receptor t-cell therapy: best practice recommendations of the European Society for Blood and Marrow Transplantation (EBMT) and the Joint Accreditation Committee of ISCT and EBMT (JACIE). Haematologica. 2020;105:297–316.
Kansagra AJ, Frey NV, Bar M, Laetsch TW, Carpenter PA, Savani BN, et al. Clinical Utilization of Chimeric Antigen Receptor T-Cells (CAR-T) in B-Cell Acute Lymphoblastic Leukemia (ALL)-an Expert Opinion from the European Society for Blood and Marrow Transplantation (EBMT) and the American Society for Blood and Marrow Transplantation (ASBMT). Bone Marrow Transpl. 2019;54:1868–80.
Park JH, Romero FA, Taur Y, Sadelain M, Brentjens RJ, Hohl TM, et al. Cytokine release syndrome grade as a predictive marker for infections in patients with relapsed or refractory B-cell acute lymphoblastic leukemia treated with chimeric antigen receptor T cells. Clinical Infect Dis. 2018;67:533–40.
Hill JA, Li D, Hay KA, Green ML, Cherian S, Chen X, et al. Infectious complications of CD19-targeted chimeric antigen receptor-modified T-cell immunotherapy. Blood. 2018;131:121–30.
Little JS, Aleissa MM, Beluch K, Gonzalez-Bocco IH, Marty FM, Manne-Goehler J, et al. Low incidence of invasive fungal disease following CD19 chimeric antigen receptor t-cell therapy for non-hodgkin lymphoma. Blood Adv. 2022;6:4821–30.
Cheok KPL, Farrow A, Springell D, O'Reilly M, Morley S, Stone N, et al. Mucormycosis after CD19 Chimeric Antigen Receptor T-Cell Therapy: Results of a US Food and Drug Administration Adverse Events Reporting System Analysis and a Review of the Literature. Lancet Infect Dis. 2024;24:e256–65.
Garner W, Palash S, Haidar G. Invasive fungal infections after anti-cd19 chimeric antigen receptor-modified t-cell therapy: state of the evidence and future directions. J Fungi. 2021;7:156.
Groll AH, Pana D, Lanternier F, Mesini A, Ammann RA, Averbuch D, et al. 8th European Conference on Infections in Leukaemia: 2020 guidelines for the diagnosis, prevention, and treatment of invasive fungal diseases in paediatric patients with cancer or post-haematopoietic cell transplantation. Lancet Oncol. 2021;22:e254–69.
Lehrnbecher T, Groll AH, Cesaro S, Alten J, Attarbaschi A, Barbaric D, et al. Invasive fungal diseases impact on outcome of childhood ALL – an analysis of the international trial AIEOP-BFM ALL 2009. Leuk 2022 371. 2022;37:72–8.
Groll AH, Abdel-Azim H, Lehrnbecher T, Steinbach WJ, Paschke A, Mangin E, et al. Pharmacokinetics and safety of posaconazole intravenous solution and powder for oral suspension in children with neutropenia: an open-label, sequential dose-escalation trial. Int J Antimicrob Agents. 2020;56:106084.
Groll AH, Körholz K, Holterhus M, Lehrnbecher T. New and emerging options for management of invasive fungal diseases in paediatric patients. Mycoses. 2024;67:e13654.
Noxafil | European Medicines Agency (EMA). Accessed November 3, 2024. https://www.ema.europa.eu/en/medicines/human/EPAR/noxafil.
Dvorak CC, Fisher BT, Esbenshade AJ, Nieder ML, Alexander S, Steinbach WJ, et al. A randomized trial of caspofungin vs triazoles prophylaxis for invasive fungal disease in pediatric allogeneic hematopoietic cell transplant. J Pediatr Infect Dis Soc. 2021;10:417–25.
Hoenigl M, Sprute R, Egger M, Arastehfar A, Cornely OA, Krause R, et al. The antifungal pipeline: fosmanogepix, ibrexafungerp, olorofim, opelconazole, and rezafungin. Drugs. 2021;81:1703–29.
Thompson GR 3rd, Soriano A, Cornely OA, Kullberg BJ, Kollef M, Vazquez J, et al. Rezafungin versus caspofungin for treatment of candidaemia and invasive candidiasis (ReSTORE): a multicentre, double-blind, double-dummy, randomised phase 3 trial. Lancet. 2023;401:49–59.
xxx. https://clinicaltrials.gov/study/NCT04368559?cond=rezafungin&rank=7.
El Ayoubi LW, Allaw F, Moussa E, Kanj SS. Ibrexafungerp: A narrative overview. Curr Res Micro Sci. 2024;6:100245.
Vanbiervliet Y, Van Nieuwenhuyse T, Aerts R, Lagrou K, Spriet I, Maertens J. Review of the novel antifungal drug olorofim (F901318). BMC Infect Dis. 2024;24. 1256.
Almajid A, Bazroon A, Al-Awami HM, Albarbari H, Alqahtani I, Almutairi R, et al. Fosmanogepix: the novel anti-fungal agent’s comprehensive review of in vitro, in vivo, and current insights from advancing clinical trials. Cureus. 2024;16:e59210.
Cornely OA, Ostermann H, Koehler P, Teschner D, Limburg E, Kramer WG, et al. Phase 1b safety and pharmacokinetics of intravenous and oral fosmanogepix in patients with acute myeloid leukaemia and neutropenia. Antimicrob Chemother. 2023;78:2645–52.
Neoh CF, Jeong W, Kong DCM, Beardsley J, Kwok P, Slavin MA, et al. New and emerging roles for inhalational and direct antifungal drug delivery approaches for treatment of invasive fungal infections. Expert Rev Anti Infect Ther. 2024;22:1085–98.
Acknowledgements
The authors and contributors thank the group GL-Events, Lyon, France for the organization of the meeting. A special thanks to Marcio Nucci who has actively participated in the last discussions, both before and after the presentation at ECIL.
Funding
the ECIL-10 meeting has been supported by unrestricted educational grants from Basilea, Gilead Sciences, MSD, Mundipharma, Takeda, F2G, Shionogi, Moderna, OLM, AstraZeneca, Scynexis and Pfizer.
Author information
Authors and Affiliations
Consortia
Contributions
All co-authors prepared a set of summary slides on each of the substances addressed in the manuscript, presented the slides at the ECIL-10 conference and revised the slide sets after the plenary discussion. LP prepared the manuscript, all co-authors revised the text and consented the final version.
Corresponding author
Ethics declarations
Competing interests
Livio Pagano (LP) was Board member of Gilead Science, MSD, Pfizer, Menarini-Stemline, Gentili, Jazz Pharmaceutical, Cidara and has received speaker’s honoraria from Gilead Sciences, Kyowa Kirin, MSD, Pfizer Pharmaceuticals, Astellas Pharma, Novartis, Jazz Pharmaceutical, Janseen-Cilag, Astra Zeneca, F2G, Abbvie. Consultant for Menarini, Cidara. Georg Maschmeyer (GM) has received speaker’s honoraria from Gilead Sciences. Frederic Lamoth (FL) reports research funding from Gilead, MSD, Pfizer and Novartis, and honoraria for conferences or advisory boards from Gilead, MSD, Pfizer, Mundipharma and Becton-Dickinson, outside of the submitted work. All contracts were made with and fees paid to his institution (CHUV). Ola Blennow has received speaker’s honoraria from Gilead Sciences. Alienor Xhaard (AX) has received speaker’s honoraria from Merck Sharp and Dohme and Pfizer Pharmaceuticals. Manuela spadea (MS) nothing to declare. Alessandro Busca (AB) was Board member of Gilead Science, Takeda, and has received speaker’s honoraria from Gilead Kite Sciences, MSD, Pfizer Pharmaceuticals, Novartis, Jazz Pharmaceutical, Astra-Zeneca, Abbvie, Menarini Stemline, Takeda. Catherine Cordonnier (CC) has received speaker’s honoraria from Gilead Science and MSD, and consulting fees from Cidara and Mundipharma. Johan Maertens (JM) reports consulting fees from Amplyx, Cidara, Gilead, Pfizer, Scynexis, F2G, Mundipharma, Takeda, and Basilea; Honoraria for lectures from Astellas, Gilead, Basilea, Mundipharma, Takeda, MedScape, Pfizer, and Shionogi; Participation on Advisory Board from Cidara, Pulmocide, Shionogi, Basilea, Sfunga.
ECIL members who participated in the drafting of these recommendations
Livio Pagano, Georg Maschmeyer, Frederic Lamoth, Ola Blennow, Alienor Xhaard, Manuela Spadea, Alessandro Busca, Catherine Cordonnier, Johan Maertens.
Additional information
Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.
About this article
Cite this article
Pagano, L., Maschmeyer, G., Lamoth, F. et al. Primary antifungal prophylaxis in hematological malignancies. Updated clinical practice guidelines by the European Conference on Infections in Leukemia (ECIL). Leukemia 39, 1547–1557 (2025). https://doi.org/10.1038/s41375-025-02586-7
Received:
Revised:
Accepted:
Published:
Issue date:
DOI: https://doi.org/10.1038/s41375-025-02586-7