Abstract
Patients with haematological malignancies (HM) are at high risk of developing invasive fungal disease (IFD) with high morbidity and attributable mortality. We reviewed data published until September 2021 to update the 2017 antifungal prophylaxis recommendations of the German Society of Haematology and Medical Oncology (DGHO). The strong recommendation to administer antifungal prophylaxis in patients with HM with long-lasting neutropenia, i.e. <500 cells/μL for >7 days remains unchanged. Posaconazole remains the drug of choice for mould-active prophylaxis in these patients. Novel treatment options in HM, such as CAR-T-cell treatment or novel targeted therapies for acute myeloid leukaemia (AML) were considered, however, data are insufficient to give general recommendations for routine antifungal prophylaxis in these patients. Major changes regarding specific recommendations compared to the 2017 edition are the now moderate instead of mild support for the recommendations of isavuconazole and voriconazole. Furthermore, published evidence on micafungin allows recommending it at moderate strength for its use in HM. For the first time we included recommendations for non-pharmaceutical measures regarding IFD, comprising the use of high-efficiency particulate air (HEPA) filters, smoking, measures during construction work and neutropenic diets.
We reviewed the impact of antifungal prophylaxis with triazoles on drug–drug interactions with novel targeted therapies that are metabolized via cytochrome p450 where triazoles inhibit CYP3A4/5. The working group recommends reducing the dose of venetoclax when used concomitantly with strong CYP3A4 inhibiting antifungals. Furthermore, we reviewed data on the prophylactic use of novel antifungal agents. Currently there is no evidence to support their use in a prophylactic setting in clinical practice.
Introduction
Invasive fungal disease (IFD) remains an important cause of severe morbidity and high mortality in patients with haematological malignancies (HM). Patients with long-lasting neutropenia (≤500/μL for ≥7 days), such as patients with acute myeloid leukaemia (AML) or myelodysplastic syndrome (MDS) during remission-induction chemotherapy (RIC) or patients with severe aplastic anaemia, continue to represent the population at highest risk of developing IFD.1,2
The most frequently identified fungal pathogens are Aspergillus spp. and Candida spp., significantly contributing to mortality in these patients.3,4 Therefore, mould-active antifungal prophylaxis has been established as a standard-of-care in these patients while those with shorter duration of expected neutropenia (≤500/μL for <7 days) are not considered to be at increased risk of IFD.5 Other strategies, such as pre-emptive or empiric treatment for IFD are widely implemented, however, these are not discussed in this guideline.6
The fungal epidemiology has changed since implementation of routine antifungal prophylaxis with the emergence of resistant fungal pathogens and identification of novel species causing breakthrough IFD (bIFD).7
New aspects, such as increased use of targeted drugs and immunomodulating treatment approaches (e.g. chimeric antigen receptor (CAR)-T cells) in many haematological entities fuel the discussion on implementation of antifungal prophylaxis.8–10 Patients and physician face unknown effects on immune response to fungal pathogens, and in addition, cytochrome p450-mediated potential drug–drug interactions (DDI) between established antifungals and new antineoplastic approaches.9,11
Design and methods
An expert group of clinical experts in haematology, oncology, infectious diseases and stem cell transplantation of the Infectious Diseases Working Party (AGIHO) of the German Society of Haematology and Medical Oncology (DGHO) prepared this guideline document in an established consensus process from August 2021 to January 2022.
This guideline focuses on adult patients and primary antifungal prophylaxis only and excludes autologous and allogeneic HSCT patients. These patient populations as well as treatment of IFD and recommendations regarding antibiotic prophylaxis and prophylaxis of Pneumocystis jirovecii pneumonia (PJP) are discussed in separate AGIHO guidelines.5,12–14
Topics were distributed among the authors and systematic literature research in PubMed in English language journals was conducted by all authors including the search terms as previously described from August to September 2021. Full texts for all included studies were obtained. Data were extracted and tabulated. Preliminary recommendations for each antifungal agent and patient group were discussed in three online meetings between October and December 2021. Tabulated data were accessible at any time to all authors. If consensus for a recommendation could not be reached by discussion, a majority vote was adopted. The final version of this manuscript was again discussed and finally approved in the present version by the full author panel.
For the grading of quality of evidence (QoE) and strength of recommendations (SoR), established methodology by the European Society of Clinical Microbiology and Infectious Diseases (ESCMID) and the European Confederation of Medical Mycology (ECMM) was implemented (Table 1).15
Table 1.
Category, grade | Definition | |
---|---|---|
Strength of recommendation | A | Strongly supports a recommendation for use |
B | Moderate evidence to support a recommendation for use | |
C | Poor evidence to support a recommendation | |
D | Supports a recommendation against use | |
Quality of evidence—Level | I | Evidence from ≥1 properly randomized controlled trial |
II | Evidence from ≥1 well-designed clinical trial, without randomization; from cohort or case-controlled analytic studies (preferably from >1 centre); from multiple time series; or from dramatic results from uncontrolled experiments | |
III | Evidence from opinions of respected authorities, based on clinical experience, descriptive studies or reports of expert committees | |
Quality of evidence—Index (for Level II) | r | Meta-analysis or systematic review of RCT |
t | Transferred evidence, that is, results from different patients’ cohorts, or similar immune-status situation | |
h | Comparator group is a historical control | |
u | Uncontrolled trial | |
a | Published abstract (presented at an international symposium or meeting) |
We discuss changes of QoE and SoR wherever applicable compared to the previous editions of this guideline.5,16,17
The reader is also referred to an updated summary of trials on antifungal prophylaxis published to date by antifungal drug with tabulated information on authors, publishing year, trial design, medication and daily dose per treatment group, number of patients, population characteristics/risk factors, share of proven, probable and possible IFD, and mortality (Tables S1–S8, available as Supplementary data at JAC Online).
The recommendations are evidence- and consensus-based, but do not necessarily follow approved indications or the respective labelling of antifungal compounds in different countries or regions.
Results
Since the 2017 edition of this guideline, 38 novel studies comprising 5083 patients receiving primary antifungal prophylaxis have been identified and analysed.
An overview of our recommendations separated by antifungal compounds is tabulated synoptically in Tables 2 and 3.
Table 2.
Intention | Intervention | SoR | QoE |
---|---|---|---|
Prevent IFD in patients with neutropenia (<500 cells/µL >7 days), excluding allogeneic HSCT | Posaconazole | A | Ia |
B | IIIb | ||
Amphotericin B, liposomal, inhalation | B | II | |
Isavuconazole | B | IIt | |
Voriconazole | B | IIu | |
Micafungin | B | II u,t | |
Amphotericin B, liposomal, i.v. | C | I | |
Caspofungin | C | I | |
Fluconazole | C | I | |
Itraconazole, p.o. and i.v. | C | I | |
SUBA-Itraconazole | C | IIt,h | |
Amphotericin B deoxycholate | D | I |
Strong recommendation in AML/MDS RIC only.
Other settings, e.g. VSAA and palliative treatment of MDS.
HSCT, haematopoietic stem cell transplantation; AML, acute myeloid leukaemia; MDS, myelodysplastic syndrome; SUBA, SuperBioAvailability.
Table 3.
Drug | Dosage |
---|---|
Posaconazole, oral suspension | 200 mg q8h p.o. |
Posaconazole, tablet | 300 mg q24h p.o. (q12h on day 1) |
Posaconazole, i.v. | 300 mg q24h i.v. (q12h on day 1) |
Amphotericin B, liposomal, inhalation | 12.5 mg twice weekly |
Amphotericin B, liposomal, i.v. | Dosage not defined; variable dosages and dosing intervals |
Caspofungin | 50 mg q24h i.v. (70 mg on day 1, 70 mg q24h if patient weighs >80 kg) |
Micafungin | 50 mg q24h i.v. |
Anidulafungin | 100 mg q24h i.v. (200 mg on day 1) |
Fluconazole | 400 mg q24h p.o. |
Itraconazole, capsules or i.v. formulation | 200 mg q24h p.o./i.v. |
Itraconazole, oral solution | 2.5–7.5 mg/kg/d or 200 mg q24h |
SUBA-itraconazole | 200 mg q24h p.o. |
Voriconazole | 4 mg/kg q12h i.v./p.o. |
Isavuconazole | 200 mg q24h i.v. (q8h on days 1–2) |
i.v., intravenous; p.o., per os; SUBA, SuperBioAvailability.
Antifungal prophylaxis is recommended in patients with long-lasting neutropenia (<500 cells/μL for >7 days) independent from the underlying disease. This typically includes patients with AML or MDS during RIC but also patients with AML/MDS during consolidation chemotherapy, patients with ALL, aplastic anaemia or with relapsed/refractory AML/MDS having curative treatment options.18,19
In contrast, patients with shorter expected duration of neutropenia (i.e. <500 cells/μL for ≤7 days) are not at significantly increased risk to develop IFD and should not receive routine antifungal prophylaxis (DI). This comprises patients treated with CAR-T cells and after high-dose chemotherapy with autologous HSCT as well as patients with lymphoma or multiple myeloma.20
Azoles
The orally available azoles are the drugs of choice for antifungal prophylaxis. However, there are substantial differences between the various azoles in terms of antifungal spectrum, absorption and DDI. Due to its efficacy and readily absorbable oral tablet formulation the mould-active posaconazole remains the drug of choice for antifungal prophylaxis (AI).
In a network meta-analysis and pharmacoeconomic evaluation of triazole prophylaxis on 5505 participants in 21 randomized controlled trials (RCT) with HM or HSCT, excluding itraconazole capsules, all triazole antifungals were effective in reducing IFD. However, the antifungal efficacy of fluconazole was lower compared to posaconazole or voriconazole.21
Posaconazole
Evidence
In patients undergoing RIC for AML or MDS in a well-designed phase 3 RCT, posaconazole significantly reduced incidence of proven and probable IFD and all-cause mortality.19 In this trial, posaconazole oral suspension was compared to the former standards of fluconazole or itraconazole solution.19 With the development of posaconazole delayed release tablets and intravenous formulations, non-comparative phase 1b/3 studies found favourable pharmacokinetic results, i.e. drug exposure sufficient for prophylactic efficacy in most patients. No new safety signal was found, including in patients with high plasma concentrations.22–25 Of note, the intravenous formulation has a very low pH and should be administered via central venous line.24,25
Since the 2017 edition of this guideline, three large retrospective cohort studies have reported results in line with the prospective studies mentioned before. A US study compared oral suspension and delayed release tablet formulations in 547 consecutive patients with AML (69%), graft-versus-host disease (GvHD) (18%) or MDS (3%). The incidence of proven or probable bIFD was 1.6% and did not differ significantly between posaconazole formulations. In eight of these 14 bIFD serum concentrations were determined, and in 7 of 8 they were ≥0.7 µg/mL.26 A retrospective study from Spain compared prophylaxis with posaconazole oral suspension with intravenous itraconazole in 174 consecutive patients treated for AML or MDS. Rates of proven or probable bIFD were 1.7% and 5.3%.27 A study from South Korea compared posaconazole prophylaxis with no prophylaxis in 247 patients with AML. Incidence rates of proven or probable IFD were 2.5% and 9.4%.28
Recommendation
AGIHO strongly recommends posaconazole prophylaxis for patients undergoing RIC for AML or MDS (AI). The previous recommendation for very severe aplastic anaemia (VSAA) and less intensive treatment settings for AML/MDS remains unchanged (B III) due to a lack of well-designed prospective studies in this specific population and treatment setting.
The formulations appear interchangeable and can be chosen according to the individual patient situation and preference. Posaconazole infusion should be considered in patients who are unable to swallow an oral drug.
Fluconazole
Evidence
Since 2018, one prospective study on fluconazole prophylaxis was conducted. In this multi-centre, randomized, open-label trial caspofungin versus fluconazole was compared for prophylaxis in children, adolescents and young adults with newly diagnosed de novo, relapsed or secondary AML during neutropenia. The 5-month cumulative incidence of IFD was 3.1% in the caspofungin group versus 7.2% in the fluconazole group, and 0.5% versus 3.1% for invasive aspergillosis. In this population, prophylaxis with caspofungin compared with fluconazole resulted in significantly lower incidence of IFD, although limited due to the paediatric setting.29
Recommendation
Our recommendation regarding fluconazole prophylaxis in long-term neutropenic haematology patients remains low (CI).
Isavuconazole
Evidence
Isavuconazole has been evaluated for primary or secondary antifungal prophylaxis in several retrospective and prospective studies. Hereby, most studies were focused on patients after HSCT30,31 or solid organ transplantation.32 More limited data are available on isavuconazole antifungal prophylaxis in non-transplant patients with haematological diseases.33–35 Efficacy and tolerability of prophylactic isavuconazole in comparison to other antifungal agents such as posaconazole or voriconazole still remains controversial. Bogler et al. performed a propensity score matched cohort analysis including allogeneic HSCT recipients of whom 210 received voriconazole and 95 isavuconazole antifungal prophylaxis. While efficacy did not differ significantly between both study groups (incidences of bIFD at day 180 were 2.9% and 3.2%, respectively), isavuconazole was better tolerated than voriconazole.30 By contrast, another retrospective study including 145 patients with haematological diseases with or without previous HSCT who received 197 courses of isavuconazole prophylaxis found that isavuconazole prophylaxis was associated with a higher percentage of bIFD compared to either posaconazole or voriconazole.34 Here, bIFD occurred in 10.2% of isavuconazole, 4.1% of posaconazole and 1.1% of voriconazole courses among patients with de novo or relapsed/refractory AML.
Recommendation
Isavuconazole might be considered as primary or secondary antifungal prophylaxis in long-term neutropenic haematology patients (BIIt).
Voriconazole
Evidence
Voriconazole remains a cornerstone in the treatment of aspergillosis and some other invasive mould infections.36,37 Results for voriconazole in the prophylaxis setting had been less convincing, with the largest studies conducted in the early phase after allogeneic HSCT38. Several retrospective studies have recently evaluated voriconazole in patients with acute leukaemia receiving chemotherapy.34,39–45 One retrospective study (n = 175) found an incidence of bIFD of 3.3% in the voriconazole arm versus 7.2% in the fluconazole arm.39 In another study in 241 AML patients receiving (re)induction chemotherapy, bIFD rate was 1.1% in the voriconazole arm, slightly lower than observed with either isavuconazole or posaconazole prophylaxis.34 A study in AML/MDS patients found that a switch to intravenous antifungals was significantly less common in those 471 patients receiving voriconazole prophylaxis (20.6%) versus those receiving fluconazole or itraconazole (30.1%).43
Recommendation
Our recommendation for antifungal prophylaxis with voriconazole in neutropenic haematology patients was upgraded to BIIu.
Itraconazole
Evidence
No prospective clinical trials investigating itraconazole for antifungal prophylaxis in HM have been published since 2018. One retrospective, single-centre, observational study comparing posaconazole (n = 179) with itraconazole in the prevention of IFD in AML/MDS patients during intensive chemotherapy showed statistically significant differences in the rates of proven or probable IFD (1.4% versus 5.3%).27 Another retrospective, single-centre, observational study comparing posaconazole (n = 45) with itraconazole (n = 90) in the prevention of IFD during AML RIC yielded similar results (bIFD rate 2.2% versus 5.5%).46
One single-centre prospective cohort study compared the use of Super BioAvailability (SUBA®)-itraconazole (n = 27) versus a historical control group of conventional liquid itraconazole (n = 30) for prophylaxis in patients with HM or undergoing allogeneic HSCT, achieving faster and more stable serum through concentrations.47
One non-comparative retrospective study (n = 74) evaluated safety and tolerability of SUBA-itraconazole, showing moderate rates of breakthrough proven/probable IFD (6%).48 Another single-centre, retrospective study in lung transplant patients (n = 150) compared triazoles (n = 78) with SUBA-itraconazole (n = 88), with equal incidence rates of IFD (two per group, respectively). However, this study was not designed to assess the efficacy of antifungal prophylaxis.49
Recommendation
The AGIHO recommendation for itraconazole or SUBA®-itraconazole prophylaxis for neutropenic haematology patients remains low (CI and CIIt, h, respectively).
Other azoles
Evidence
For other azoles, no relevant literature regarding prophylaxis in HM has been published since 2017. One meta-analysis to assess prevention of oral candidiasis showed the lowest rates for oral candidiasis in patients with clotrimazole treatment, however, it had no comparative studies and did not assess systemic IFD.50 There is no evidence to support the prophylactic use of clotrimazole, miconazole or ketoconazole (DII).
Recommendation
There is a recommendation against the use of other than the previously listed azole antifungals for systemic antifungal prophylaxis (DII).
Echinocandins
Echinocandins are mostly used for treatment of candidemia reducing overall attributable mortality.51 However, the use of echinocandins as first-line antifungal prophylaxis is not recommended due to limited evidence from RCTs in this setting.
Anidulafungin
Since 2017, no additional studies have been published for anidulafungin or caspofungin as antifungal prophylaxis in adults.
Caspofungin
Since 2017, one multi-centre, randomized, open-label trial compared caspofungin versus fluconazole for prophylaxis in children, adolescents and young adults with newly diagnosed de novo, relapsed or secondary AML during neutropenia following induction and consolidation chemotherapy. Among the 517 randomized participants, 22 in the caspofungin arm and 18 in the fluconazole arm were 18 years or older (median age 9 years). Twenty-three proven or probable IFD events (six in the caspofungin group versus 17 in the fluconazole group) including 14 moulds, seven yeasts and two fungi not further speciated were detected. The 5-month cumulative incidence of IFD was 3.1% in the caspofungin group versus 7.2% in the fluconazole group, and for invasive aspergillosis, it was 0.5% with caspofungin versus 3.1% with fluconazole. In this study, prophylaxis with caspofungin resulted in a significantly lower incidence of IFD; however, the reduction in the very small adult population (n = 40, 7.7%) in this trial was not determined and is therefore not fully considered in this guideline.29 The prophylactic use of caspofungin has also been shown to be non-inferior to triazole prophylaxis in another RCT in the paediatric allogeneic HSCT population.52
Micafungin
Evidence
Several retrospective studies53–58 in the transplant setting (allogeneic HSCT and SOT) as well as one prospective clinical trial59 assessing micafungin prophylaxis in the non-transplant setting have been published since the last update of the guideline. The prospective trial included patients undergoing RIC for AML who received micafungin once daily from the first day of induction therapy until the end of neutropenia.59 None of the 41 patients developed bIFD. Further retrospective studies in the transplant setting confirmed these findings in larger sample sizes (ranging from 69 to 216 included patients).
Recommendation
Considering the small sample size of the prospective study as well as retrospective data from allogeneic HSCT and SOT patients, our recommendation for micafungin in neutropenic HM changes from CIIh to BIIt, u and the recommendation for caspofungin remains unchanged (CI) due to very limited data in the adult population whereas anidulafungin does not receive a recommendation due to lack of evidence.
Polyenes
Evidence
Regarding the emerging threat of increasingly detected azole-resistant isolates, non-azole antifungal drugs may be of importance for future prophylactic strategies. Comparable to posaconazole, liposomal amphotericin B (L-AmB) exhibits broad-spectrum activity and thus may be helpful in a prophylactic setting. The use of polyenes has been studied in different populations and several clinical trials. Intravenous (IV) L-AmB prophylaxis has been evaluated in adult ALL patients during RIC.18 The choice of L-AmB in this specific setting arises from CYP3A4-mediated DDI of azoles with vinca-alkaloids that prohibit the concomitant use of azoles during chemotherapy. However, there was no significant difference in IFD incidence comparing L-AmB 5 mg/kg per week and placebo recipients (7.9% versus 11.7%).18 There is poor evidence to recommend IV L-AmB prophylaxis in ALL (CI). Several other dosing regimens have been used in clinical studies, e.g. a standard dose of 50 mg/q48h60 or, in the most recent studies, weight-adapted regimens such as 1 mg/kg three times weekly, 3 mg/kg weekly up to 7.5 mg/kg weekly.18,61–63 Safety and efficacy of these dosing regimens have not been compared systematically, therefore we refrain from recommending a specific dose.
The prophylactic use of aerosolized L-AmB in severely neutropenic patients was graded with BII in the previous versions of this guideline5,16 as it significantly reduced invasive pulmonary aspergillosis rates and was cost efficient.64,65
Recommendation
Our recommendation for the prophylactic use of IV L-AmB with any dose in neutropenic HM remains unchanged (CI). The recommendation for aerosolized L-AmB remains BII; note that this should be administered concomitantly to systematic fluconazole for prophylaxis of candidemia. L-AmB prophylaxis may play an important role in centres with higher rates of azole-resistant fungal isolates. However, the group recommends against the use of amphotericin B deoxycholate due to its toxicity (DI).
Other antifungals (nystatin, terbinafine)
A comprehensive literature review from 2014 identified a meta-analysis from 11 historic trials in cancer patients where nystatin was used as antifungal prophylaxis.66 No benefit compared to a placebo was found and nystatin is not recommended in this indication (DIIr).
Otherwise, no additional literature has been published since 2017. There is no evidence to support the prophylactic use of terbinafine (DII).
Therapeutic drug monitoring and metabolism
Evidence
Therapeutic drug monitoring (TDM) of antifungals may be useful as toxicity depends on plasma drug levels and inter- and intraindividual pharmacokinetics may vary. However, the association of triazole plasma concentration and efficacy has primarily been shown in the setting of IFD treatment and for itraconazole.67 Prospective studies proving a plasma concentration-dependent effect on clinical outcome or adverse events in the setting of prophylaxis are scarce.68 However, the results of some retrospective analyses indicate which azole levels may be required to protect against IFD and avoid toxicity.
Recommendation
In general, assessment of plasma concentration is recommended for triazoles in case of a (suspected) bIFD (AIII) to understand potential reasons for IFD and scope treatment options. In addition, TDM of specific azoles may be useful in specific clinical situations where resorption or metabolism might be affected, e.g. in obesity, renal/organ replacement therapy, gastrointestinal GvHD or intensive care (CIIt).69 Recommendations for specific triazoles are listed in Table 4. To establish TDM, blood samples should be drawn 3 days (for posaconazole and voriconazole) or 7 days (for itraconazole) after initiation and dose adjustment of antifungal triazole prophylaxis or change of interfering medication.70 TDM is not well established for fluconazole, echinocandins and polyenes and is therefore not recommended.
Table 4.
Drug | Rationale | Target | SoR | QoE | Comment | Reference |
---|---|---|---|---|---|---|
Any triazole: in case of suspected breakthrough IFD |
To clarify treatment options | Variable (see below) | A | III | ||
Oral itraconazole | To monitor for efficacy and toxicity | >0.5 mg/L | B | IIt | 104–107 | |
Isavuconazole | To monitor in case of toxicity | 2–5 mg/L | C | IIt | Higher concentrations have been associated with an increased risk of adverse events | 33,68,108–113 |
Posaconazole oral suspension | To support efficacy; in case of suspected impaired resorption | >0.7 mg/L (prophylaxis) > 1 mg/L (treatment) | B | IIt | Reduced plasma levels have been demonstrated e.g. in case of GI-GvHD, diarrhoea, concomitant PPI | 19,114–125 |
Posaconazole oral or i.v. |
To support efficacy | B | III | |||
Voriconazole | To support efficacy | >1 mg/L | B | IIt | 126,127 | |
Voriconazole | To avoid toxicity | <4.5 mg/L | A | II | Recommendation in case of clinically attributed toxicity |
Comment: recommendations are not generally applicable for a prophylactic setting and refer to specific situation, see section ‘Therapeutic drug monitoring and metabolism’.
GI-GvHD, gastrointestinal graft-versus-host-disease; IFD, invasive fungal infection; PPI, proton pump inhibitors
Antifungal prophylaxis, targeted therapies and potential drug–drug interactions
Evidence
Targeted antineoplastic therapy for AML is fraught with uncertainties regarding pharmacokinetic compatibility with antifungal prophylaxis, especially strong CYP3A4 inhibitors, with in vitro data suggesting potential DDI, however, clinical data on the impact of potential DDI remain sparse (Table S9).71
The quantitatively most important and well-studied DDI exists for triazole antifungals and the bcl-2 inhibitor venetoclax. A PK study of 12 patients with AML determined the need to reduce venetoclax dose by at least 75% in combination with posaconazole to achieve equivalent serum levels compared to venetoclax monotherapy.72 The determination of the exact dose of venetoclax is an ongoing debate and clinical trials are continuing.73 A retrospective cohort study analysed 121 AML patients treated with venetoclax and hypomethylating agents, 89 of theese concomitantly received an azole.74,75 The combination resulted in prolonged cytopenia without increased rates of febrile neutropenia, infections or duration of hospitalization. Omission of venetoclax dose reduction was associated with numerically higher rates of these complications. The duration of antifungal prophylaxis in patients receiving venetoclax should be guided by neutropenia; note that the venetoclax dose must be increased on termination of moderate and strong CYP3A4 inhibitors.71 Bose et al. reported no increase of isavuconazole serum levels or associated toxicities in a cohort of 65 AML patients receiving primary isavuconazole prophylaxis during RIC, 27 of which concomitantly received venetoclax alone or in combination with an FLT3-inhibitor.33
A retrospective analysis of midostaurin with concomitant strong CYP3A4 inhibitors, including posaconazole and voriconazole, from the phase III RATIFY trial demonstrated an earlier onset of but no overall increase in adverse events.76
For the second generation FLT3-inhibitor gilteritinib, no significant difference in toxicities, need for dose reduction or clinical outcomes was reported between patients with AML receiving or not receiving concomitant triazole prophylaxis.77
Recommendation
The guideline group strongly recommends reducing the dose of venetoclax by at least 75% when administered concomitantly to strong CYP3A4 inhibitors (AI). For all other novel targeted therapies, well-designed studies with combined clinical and pharmacokinetic endpoints are currently lacking (Table 5).
Table 5.
Population | Intention | Intervention | SoR | QoE | Reference |
---|---|---|---|---|---|
AML/MDS patients treated with | |||||
Venetoclax | Prevent IFD | use triazole antifungal prophylaxis | Aa | IIu,t | 33, 72, 74, 75, 78 |
Prevent toxicity | Reduce dose of venetoclax by at least 75% in combination with posaconazole or voriconazole and by 50% in combination with fluconazole or isavuconazole | A | IIu,t | ||
Gilteritinib | Prevent IFD | Use triazole antifungal prophylaxis without dose adjustment | Aa | IIu | 77 |
Midostaurin | Prevent IFD | If indicated, use triazole antifungal prophylaxis without dose adjustment | A | IIu | 76 |
Quizartinib | Prevent IFD | If indicated, use triazole antifungal prophylaxis without dose adjustment | Aa | IIu,t | 79 |
Prevent toxicity | Reduce quizartinib dose (60 to 30 mg or 30 to 20 mg) in combination with posaconazole or voriconazole | B | III | ||
Ivosidenib | Prevent IFD | If indicated, use triazole antifungal prophylaxis without dose adjustment | Aa | III | 80 |
Prevent toxicity | Reduce ivosidenib dose to 250 mg/day in combination with posaconazole or voriconazole | B | III |
AML, acute myeloid leukaemia; IFD, invasive fungal disease; MDS, myelodysplastic syndrome.
Strong recommendation for antifungal prophylaxis, if neutropenia ≥7 days is expected or present.
Novel antifungals
Several new antifungal classes in late-stage clinical development have the potential for prophylactic use (Table 6).81 Opelconazole is a novel triazole that is optimized for inhalation to maximize local efficacy while avoiding systemic toxicity.82 A phase IIb trial will investigate the prophylactic use in lung transplant recipients. Rezafungin, an echinocandin with an extended half-life and once-weekly intravenous administration is currently being evaluated in a phase 3 trial for its potential to prevent IFD by Candida spp., Aspergillus spp. and P. jirovecii in allogeneic HSCT recipients (NCT04368559).83 Its prophylactic use could overcome current multidrug regimens. However, to date, clinical trial data on prophylaxis are not available for these promising novel antifungals.
Table 6.
Antifungal | Mechanism of action | Future areas of use | Future use in prophylaxis | Clinical trials evaluating prophylactic use |
---|---|---|---|---|
Fosmanogepix/Manogepix | Inhibition of Gwt1, targets GPI-anchored protein maturation | Invasive infections with Aspergillus spp., Scedosporium spp., Fusarium spp., Mucorales, Cryptococcus spp., Candida spp. (except C. krusei) Endemic mycoses, including coccidioidomycoses |
unclear | |
Ibrexafungerp | Glucan synthase inhibitor with alternative binding site | Invasive candidiasis including C. auris and C. glabrata, resistant invasive pulmonary aspergillosis, other invasive fungal infections | PJP prophylaxis | Preclinical data |
Olorofim | Inhibition of dihydroorotate dehydrogenase, targets pyrimidine synthesis | Invasive infections with multi-resistant moulds, including resistant Aspergillus spp. and L. prolificans Endemic mycoses, including coccidioidomycoses |
Mould prophylaxis | NCT02856178 |
Opelconazole | Triazole with inhaled administration, targets lanosterol-14alpha-demethylase | Infections with Aspergillus spp. | Prophylaxis in lung transplants, ICU setting | NCT05037851 |
Rezafungin | Echinocandin with prolonged half-life, targets glucan synthase | Invasive infections with Candida spp., Aspergillus spp., Pneumocystis jirovecii | Prophylaxis in HSCT and SOT | NCT04368559 |
GPI, Glycosylphosphatidylinositol; HSCT, hematopoietic stem cell transplantation; ICU, intensive care unit; PjP, Pneumocystis jirovecii pneumonia; SOT, solid organ transplantation.
Non-pharmaceutical interventions
Non-pharmaceutical interventions were not extensively reviewed in previous versions of this guideline, thus, in this update, we decided to include it for reasons of completeness and relevance, but only included the most recent studies on this topic (Table 7).
Table 7.
Intention | Intervention | SoR | QoE | Reference |
---|---|---|---|---|
To prevent IFD | Neutropenic diet | D | IIr,u | 84–86 |
To prevent invasive aspergillosis | Wearing well-fitting (FFP2) masks | C | IIt | 87 |
To prevent IFD | HEPA filters LAF systems |
A B |
IIu IIu |
88–91 |
To prevent CVC-related fungal bloodstream infections | Chlorhexidine-coated CVC dressings | C | I | 92 |
To prevent IFD | romyelocel-L* |
B | I | 93–95 |
granulocyte transfusions | B | IIr | ||
G-CSF | B | IIu | ||
To prevent IFD | Quit smoking | A | IIu | 96–98 |
CVC, central venous catheter; FFP2, filtering face piece 2; G-CSF, granulocyte-colony-stimulating factor; HEPA, high efficiency particulate air; IFD, invasive fungal disease; LAF, laminar air flow.
*Cryopreserved human allogeneic myeloid progenitor cells.
Evidence
Filamentous fungi are ubiquitous environmental organisms, and the risk of exposure depends on various conditions, for example geography, occupation and weather, including humidity, temperature and wind. Inhalation is the most common route, but fungal uptake may occur following consumption of contaminated products or direct inoculation, too.99 Regardless of scientific evidence, recommendations of regulatory authorities should be considered, especially for patients with HSCT.100
Recommendation
In high-risk neutropenic patients, germ-free diet to minimize pathogen exposure is not beneficial for the prevention of IFD (DIIr, u), but is associated with a higher incidence of nausea, diarrhoea and weight loss.84–86
In patients with chemotherapy or HSCT for acute leukaemia, a multicentric RCT failed to prevent the occurrence of IA by wearing well-fitting face masks (CIIt).87
HEPA filters—permanent, or portable in case of construction work—(AIIu) and/or laminar air flow (LAF) systems (BIIu) are effective to prevent IFD in patients with chemotherapy for acute leukaemia.88–91
In neutropenic patients, chlorhexidine-coated CVC dressings are not recommended for prevention of CVC-related bloodstream infections, including fungemia (CI).92
Application of romyelocel-L (cryopreserved human allogeneic myeloid progenitor cells) (BI), granulocyte transfusions (BIIr) or G-CSF (BIIu) may be effective for prevention of fungal infections, but did not show survival benefits.93–95
Smoking is a risk factor for invasive pulmonary aspergillosis, independent of antifungal prophylaxis.96–98 Giving up smoking can be a patient’s personal preventive measure (AIIu).
Measuring airborne fungal concentrations, mechanical preventive measures (air lock chambers, sealed windows, surgical masks for neutropenic patients) during hospital constructions and outbreaks are important measures. However, published and unpublished evidence is contradictory, which is why the group decided to not give a graded recommendation.101,102
Discussion and conclusion
In this updated guideline, the evidenced-based recommendation for antifungal prophylaxis in patients with AML and MDS after RIC is still valid (AI).
Major changes regarding specific recommendations are an upgrade for the prophylactic use of voriconazole in neutropenic haematology patients from C to B, as more studies showing lower bIFD rates compared to other triazoles. Isavuconazole was also upgraded from C to B with more evidence from retrospective studies published in the recent years. However, with still higher bIFD rates compared to posaconazole and voriconazole. Micafungin at a dose of 50 mg per day is now recommended at a moderate strength with more evidence transferred from the allogeneic HSCT population.
Prophylaxis should be administered preferably with mould-active azoles or an echinocandin, whereby posaconazole remains the drug of choice due to its efficacy and readily absorbable oral tablet formulation (AI). In a network meta-analysis and pharmacoeconomic evaluation of triazole prophylaxis on 5505 participants in 21 RCTs with HM or HSCT, other than itraconazole capsule, all triazole antifungals were effective in reducing IFD. However, the antifungal efficacy of fluconazole was lower compared to posaconazole or voriconazole.21 In addition to the respective licensing status and the increased interaction potential, it is important to note that TDM may help monitoring potential toxicity, especially during prophylaxis with voriconazole (AIIt). Safety of voriconazole was inferior when compared to posaconazole in retrospective studies.40,45 Patients with persistent neutropenia due to active underlying malignant disease and thus an increased risk of IFD may also benefit from antifungal prophylaxis (BIII). In individual cases, the specific cellular immune status must be considered, which, in addition to new antineoplastic compounds, is the primary driver of the IFD risk.71,103 Under certain circumstances, non-pharmaceutical measures may help to prevent IFD in neutropenic haematological patients. With IFD rates remaining low in patients after high-dose chemotherapy with autologous HSCT or CAR-T-cell therapy, no general prophylaxis is recommended. For patients during or after allogeneic HSCT, we refer to the specific guideline of our society.13
Supplementary Material
Acknowledgements
We thank Muriel Rolfes and Sebastian Rahn for logistical support.
Contributor Information
Jannik Stemler, University of Cologne, Faculty of Medicine and University Hospital Cologne, Department I of Internal Medicine, Excellence Center for Medical Mycology (ECMM), Cologne, Germany; University of Cologne, Faculty of Medicine and University Hospital Cologne, Translational Research, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne, Germany; German Centre for Infection Research (DZIF), Partner Site Bonn-Cologne, Cologne, Germany.
Sibylle C Mellinghoff, University of Cologne, Faculty of Medicine and University Hospital Cologne, Department I of Internal Medicine, Excellence Center for Medical Mycology (ECMM), Cologne, Germany; University of Cologne, Faculty of Medicine and University Hospital Cologne, Translational Research, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne, Germany; German Centre for Infection Research (DZIF), Partner Site Bonn-Cologne, Cologne, Germany.
Yascha Khodamoradi, Department of Internal Medicine, Infectious Diseases, University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt am Main, Germany.
Rosanne Sprute, University of Cologne, Faculty of Medicine and University Hospital Cologne, Department I of Internal Medicine, Excellence Center for Medical Mycology (ECMM), Cologne, Germany; University of Cologne, Faculty of Medicine and University Hospital Cologne, Translational Research, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne, Germany; German Centre for Infection Research (DZIF), Partner Site Bonn-Cologne, Cologne, Germany.
Annika Y Classen, University of Cologne, Faculty of Medicine and University Hospital Cologne, Department I of Internal Medicine, Excellence Center for Medical Mycology (ECMM), Cologne, Germany; German Centre for Infection Research (DZIF), Partner Site Bonn-Cologne, Cologne, Germany.
Sonja E Zapke, Department Hematology, Oncology, Infectious disease and Palliatve Care, Helios University Hospital Wuppertal, Wuppertal, Germany.
Martin Hoenigl, Division of Infectious Diseases, Department of Internal Medicine, Excellence Center for Medical Mycology (ECMM), Medical University of Graz, Graz, Austria and BioTechMed, Graz, Austria.
Robert Krause, Division of Infectious Diseases, Department of Internal Medicine, Excellence Center for Medical Mycology (ECMM), Medical University of Graz, Graz, Austria and BioTechMed, Graz, Austria.
Martin Schmidt-Hieber, 2nd Medical Clinic (Hematology, Oncology, Pneumology, Nephrology), Carl-Thiem Clinic Cottbus, Cottbus, Germany.
Werner J Heinz, Medical Clinic II, Caritas Hospital, Bad Mergentheim, Germany.
Michael Klein, Department of Hematology and Medical Oncology, Klinikum Vest, Knappschaftskrankenhaus, Recklinghausen, Germany.
Philipp Koehler, University of Cologne, Faculty of Medicine and University Hospital Cologne, Department I of Internal Medicine, Excellence Center for Medical Mycology (ECMM), Cologne, Germany; University of Cologne, Faculty of Medicine and University Hospital Cologne, Translational Research, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne, Germany.
Blasius Liss, Department Hematology, Oncology, Infectious disease and Palliatve Care, Helios University Hospital Wuppertal, Wuppertal, Germany; School of Medicine, Faculty of Health, Witten/Herdecke University, Witten, Germany.
Michael Koldehoff, Department of Bone Marrow Transplantation, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany; Department of Hygiene and Environmental Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany.
Christoph Buhl, Medical Clinic III, Hospital Leverkusen, Leverkusen.
Olaf Penack, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Berlin, Germany; Humboldt-Universität zu Berlin, Department of Hematology, Oncology and Tumorimmunology, Berlin, Germany.
Georg Maschmeyer, Formerly Department of Hematology, Oncology and Palliative Care, Klinikum Ernst von Bergmann, Potsdam, Germany.
Enrico Schalk, Department of Haematology and Oncology, Medical Centre, Otto-von-Guericke University Magdeburg, Magdeburg, Germany.
Cornelia Lass-Flörl, Institute of Hygiene and Medical Microbiology, ECMM Excellence Centre, Medical University of Innsbruck, Innsbruck, Austria.
Meinolf Karthaus, Department of Hematology, Oncology and Palliative Care, Klinikum Neuperlach, Munich, Germany.
Markus Ruhnke, Helios Klinikum Aue, Klinik für Hämatologie/Onkologie & Palliativmedizin, Aue, Germany.
Oliver A Cornely, University of Cologne, Faculty of Medicine and University Hospital Cologne, Department I of Internal Medicine, Excellence Center for Medical Mycology (ECMM), Cologne, Germany; German Centre for Infection Research (DZIF), Partner Site Bonn-Cologne, Cologne, Germany; University of Cologne, Faculty of Medicine and University Hospital Cologne, Chair Translational Research, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne, NRW, Germany; University of Cologne, Faculty of Medicine and University Hospital Cologne, Clinical Trials Centre Cologne (ZKS Köln), Cologne, Germany.
Daniel Teschner, Department of Hematology, and Medical Oncology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany; Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany.
Funding
This work was carried out as part of our routine duties and within our voluntary work for the German Society of Haematology and Oncology and did not receive any additional funding.
Transparency declarations
J.St. has received research grants by the Ministry of Education and Research (BMBF) and Basilea Pharmaceuticals Inc.; has received speaker honoraria by Pfizer Inc., Gilead and Abbvie; has been a consultant to Gilead, Produkt&Markt GmbH, Alvea Vax. and Micron Research, and has received travel grants by German Society for Infectious Diseases (DGI e.V.) and Meta-Alexander Foundation. S.C.M. has received research support from Deutsches Zentrum für Infektionsforschung and Octapharma, has received consulting fees from Octapharma and has received travel grants from Gilead. Y.K. has licences with Elsevier, has received speaker honoraria from MSD Merck Sharp & Dohme, Ferring, Gilead Sciences, has received travel grants from Gilead Sciences, ViiV Healthcare and has participated on the data safety monitoring board of Gilead Sciences, ViiV Healthcare, MSD Merck Sharp & Dohme. R.S. has received speaker honoraria by Pfizer. A.Y.C. has no relevant conflicts of interest with regard to the topic of this guideline. M.H. has received research funding from MSD, Astellas, Pfizer, Scynexis, F2G, Euroimmun, Mundipharma, Gilead and NIH. R.K. received research grants from Merck and Pfizer and received speaker honoraria from Pfizer, Gilead, Astellas, Basilea, Merck, Angelini and Shionogi. M.S.H. has received travel grant from the German Society of Haematology and Medical Oncology (DGHO). W.J.H. has received lecture honoraria from Abbvie, Amgen, AstraZeneca, Celgene/Bristol-Myers Squibb, Gliead Sciences, Janssen, MSD Sharp & Dohme, Pfizer; has received support to attend meetings and travel support from Abbvie, Alexion, Astellas, Janssen, Lilly, MSD Sharp & Dohme, Novartis and Pfizer; and has participated on advisory boards from Abbvie, Amgen, AstraZeneca, Basilea, Celgene/BMS, Gliead Sciences, Janssen and Sanofi-Aventis. P.K. reports grants or contracts from German Federal Ministry of Research and Education (BMBF) B-FAST (Bundesweites Forschungsnetz Angewandte Surveillance und Testung) and NAPKON (Nationales Pandemie Kohorten Netz, German National Pandemic Cohort Network) of the Network University Medicine (NUM) and the State of North Rhine-Westphalia; Consulting fees Ambu GmbH, Gilead Sciences, Mundipharma Resarch Limited, Noxxon N.V. and Pfizer Pharma; Honoraria for lectures from Akademie für Infektionsmedizin e.V., Ambu GmbH, Astellas Pharma, BioRad Laboratories Inc., European Confederation of Medical Mycology, Gilead Sciences, GPR Academy Ruesselsheim, HELIOS Kliniken GmbH, Lahn-Dill-Kliniken GmbH, medupdate GmbH, MedMedia, MSD Sharp & Dohme GmbH, Pfizer Pharma GmbH, Scilink Comunicación Científica SC and University Hospital and LMU Munich; Participation on an Advisory Board from Ambu GmbH, Gilead Sciences, Mundipharma Resarch Limited and Pfizer Pharma; A pending patent currently reviewed at the German Patent and Trade Mark Office; Other non-financial interests from Elsevier, Wiley and Taylor & Francis online outside the submitted work. M.Ko. has no relevant conflicts of interest with regard to the topic of this guideline. O.P. has received honoraria or travel support from Gilead, Jazz, MSD, Novartis, Pfizer and Therakos; has received research support from Incyte and Priothera; is member of advisory boards to Equillium Bio, Jazz, Gilead, Novartis, MSD, Omeros, Priothera, Shionogi and SOBI. G.M. accepted honoraria for lectures from Gilead, Bristol-Myers Squibb, Merck-Serono, AMGEN, AstraZeneca and Janssen-Cilag and a travel grant from Janssen-Cilag. E.S. received speaker honoraria from Gilead. C.L.F. reports grants, personal fees and other from Gilead Sciences, grants and other from Astellas Pharma, personal fees from Pfizer, personal fees from Merck Sharp and Dohme, personal fees from bioMérieux, personal fees from F2G and personal fees from Immy, outside the submitted work. M.Ka. has received lecture Honoria from Gilead. M.R. has received consulting fees from Mundipharma and speaker honoraria from Novartis and Gilead. O.A.C. reports grants or contracts from Amplyx, Basilea, BMBF, Cidara, DZIF, EU-DG RTD (101037867), F2G, Gilead, Matinas, MedPace, MSD, Mundipharma, Octapharma, Pfizer, Scynexis; Consulting fees from Abbvie, Amplyx, Biocon, Biosys, Cidara, Da Volterra, Gilead, IQVIA, Janssen, Matinas, MedPace, Menarini, Molecular Partners, MSG-ERC, Noxxon, Octapharma, Pardes, Pfizer, PSI, Scynexis, Seres; Honoraria for lectures from Abbott, Abbvie, Al-Jazeera Pharmaceuticals, Astellas, Gilead, Grupo Biotoscana/United Medical/Knight, Hikma, MedScape, MedUpdate, Merck/MSD, Mylan, Noscendo, Pfizer, Shionogi; Payment for expert testimony from Cidara; Participation on a Data Safety Monitoring Board or Advisory Board from Actelion, Allecra, Cidara, Entasis, IQVIA, Janssen, MedPace, Paratek, PSI, Pulmocide, Shionogi, The Prime Meridian Group; A patent at the German Patent and Trade Mark Office (DE 10 2021 113 007.7); Stocks from CoRe Consulting; Other interests from DGHO, DGI, ECMM, ISHAM, MSG-ERC and Wiley. D.T. has received research grants by Gilead; has received speaker honoraria by Gilead, MSD and Pfizer; has been a consultant to Gilead, and MSD and has received travel grants by Astellas, the German Society for Haematology and Medical Oncology (DGHO), Gilead and the Paul Ehrlich Society (PEG). All other authors declare no conflict of interest.
Supplementary data
Tables S1 to S9 are available as Supplementary data at JAC Online.
References
- 1. Pagano L, Busca A, Candoni A et al. Risk stratification for invasive fungal infections in patients with hematological malignancies: SEIFEM recommendations. Blood Rev 2017; 31: 17–29. 10.1016/j.blre.2016.09.002 [DOI] [PubMed] [Google Scholar]
- 2. Koehler P, Hamprecht A, Bader O et al. Epidemiology of invasive aspergillosis and azole resistance in patients with acute leukaemia: the SEPIA study. Int J Antimicrob Agents 2017; 49: 218–23. 10.1016/j.ijantimicag.2016.10.019 [DOI] [PubMed] [Google Scholar]
- 3. Posteraro B, De Carolis E, Criscuolo M et al. Candidaemia in haematological malignancy patients from a SEIFEM study: epidemiological patterns according to antifungal prophylaxis. Mycoses 2020; 63: 900–10. 10.1111/myc.13130 [DOI] [PubMed] [Google Scholar]
- 4. Bergamasco MD, Pereira CAP, Arrais-Rodrigues C et al. Epidemiology of invasive fungal diseases in patients with hematologic malignancies and hematopoietic cell transplantation recipients managed with an antifungal diagnostic driven approach. J Fungi (Basel) 2021; 7:588. 10.3390/jof7080588 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. Mellinghoff SC, Panse J, Alakel N et al. Primary prophylaxis of invasive fungal infections in patients with haematological malignancies: 2017 update of the recommendations of the infectious diseases working party (AGIHO) of the German Society for Haematology and Medical Oncology (DGHO). Ann Hematol 2018; 97: 197–207. 10.1007/s00277-017-3196-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Coussement J, Lindsay J, Teh BW et al. Choice and duration of antifungal prophylaxis and treatment in high-risk haematology patients. Curr Opin Infect Dis 2021; 34: 297–306. 10.1097/QCO.0000000000000737 [DOI] [PubMed] [Google Scholar]
- 7. Jenks JD, Cornely OA, Chen SC et al. Breakthrough invasive fungal infections: who is at risk? Mycoses 2020; 63: 1021–32. 10.1111/myc.13148 [DOI] [PubMed] [Google Scholar]
- 8. Haidar G, Garner W, Hill JA. Infections after anti-CD19 chimeric antigen receptor T-cell therapy for hematologic malignancies: timeline, prevention, and uncertainties. Curr Opin Infect Dis 2020; 33: 449–57. 10.1097/QCO.0000000000000679 [DOI] [PubMed] [Google Scholar]
- 9. Haidar G, Dorritie K, Farah R et al. Invasive mold infections after chimeric antigen receptor-modified T-cell therapy: a case series, review of the literature, and implications for prophylaxis. Clin Infect Dis 2020; 71: 672–6. 10.1093/cid/ciz1127 [DOI] [PubMed] [Google Scholar]
- 10. Garner W, Samanta P, Haidar G. Invasive fungal infections after anti-CD19 chimeric antigen receptor-modified T-cell therapy: state of the evidence and future directions. J Fungi (Basel) 2021; 7:156. 10.3390/jof7020156 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Kantarjian H, Short NJ, DiNardo C et al. Harnessing the benefits of available targeted therapies in acute myeloid leukaemia. Lancet Haematol 2021; 8: e922–e33. 10.1016/S2352-3026(21)00270-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Christopeit M, Schmidt-Hieber M, Sprute R et al. Prophylaxis, diagnosis and therapy of infections in patients undergoing high-dose chemotherapy and autologous haematopoietic stem cell transplantation. 2020 update of the recommendations of the infectious diseases working party (AGIHO) of the German society of hematology and medical oncology (DGHO). Ann Hematol 2021; 100: 321–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. Ullmann AJ, Schmidt-Hieber M, Bertz H et al. Infectious diseases in allogeneic haematopoietic stem cell transplantation: prevention and prophylaxis strategy guidelines 2016. Ann Hematol 2016; 95: 1435–55. 10.1007/s00277-016-2711-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Classen AY, Henze L, von Lilienfeld-Toal M et al. Primary prophylaxis of bacterial infections and Pneumocystis jirovecii pneumonia in patients with hematologic malignancies and solid tumors: 2020 updated guidelines of the infectious diseases working party of the German Society of Hematology and Medical Oncology (AGIHO/DGHO). Ann Hematol 2021; 100: 1603–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Cornely OA, Cuenca-Estrella M, Meis JF et al. European Society of Clinical Microbiology and Infectious Diseases (ESCMID) Fungal Infection Study Group (EFISG) and European Confederation of Medical Mycology (ECMM) 2013 joint guidelines on diagnosis and management of rare and emerging fungal diseases. Clin Microbiol Infect 2014; 20 Suppl 3: 1–4. 10.1111/1469-0691.12569 [DOI] [PubMed] [Google Scholar]
- 16. Tacke D, Buchheidt D, Karthaus M et al. Primary prophylaxis of invasive fungal infections in patients with haematologic malignancies. 2014 update of the recommendations of the infectious diseases working party of the German Society for Haematology and Oncology. Ann Hematol 2014; 93: 1449–56. 10.1007/s00277-014-2108-y [DOI] [PubMed] [Google Scholar]
- 17. Cornely OA, Böhme A, Buchheidt D et al. Primary prophylaxis of invasive fungal infections in patients with hematologic malignancies. Recommendations of the infectious diseases working party of the German Society for Haematology and Oncology. Haematologica 2009; 94: 113–22. 10.3324/haematol.11665 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Cornely OA, Leguay T, Maertens J 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. 10.1093/jac/dkx133 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Cornely OA, Maertens J, Winston DJ et al. Posaconazole vs. f luconazole or itraconazole prophylaxis in patients with neutropenia. N Engl J Med 2007; 356: 348–59. 10.1056/NEJMoa061094 [DOI] [PubMed] [Google Scholar]
- 20. Reich G, Cornely OA, Sandherr M et al. Empirical antimicrobial monotherapy in patients after high-dose chemotherapy and autologous stem cell transplantation: a randomised, multicentre trial. Br J Haematol 2005; 130: 265–70. 10.1111/j.1365-2141.2005.05608.x [DOI] [PubMed] [Google Scholar]
- 21. Zhao YJ, Khoo AL, Tan G et al. Network meta-analysis and pharmacoeconomic evaluation of fluconazole, itraconazole, posaconazole, and voriconazole in invasive fungal infection prophylaxis. Antimicrob Agents Chemother 2016; 60: 376–86. 10.1128/AAC.01985-15 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Cornely OA, Duarte RF, Haider S et al. Phase 3 pharmacokinetics and safety study of a posaconazole tablet formulation in patients at risk for invasive fungal disease. J Antimicrob Chemother 2016; 71: 718–26. 10.1093/jac/dkv380 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Cornely OA, Duarte RF, Haider S et al. Phase 3 pharmacokinetics and safety study of a posaconazole tablet formulation in patients at risk for invasive fungal disease. J Antimicrob Chemother 2016; 71: 1747. 10.1093/jac/dkw079 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Cornely OA, Robertson MN, Haider S et al. Pharmacokinetics and safety results from the phase 3 randomized, open-label, study of intravenous posaconazole in patients at risk of invasive fungal disease. J Antimicrob Chemother 2017; 72: 3406–13. 10.1093/jac/dkx263 [DOI] [PubMed] [Google Scholar]
- 25. Cornely OA, Robertson MN, Haider S et al. Pharmacokinetics and safety results from the phase 3 randomized, open-label, study of intravenous posaconazole in patients at risk of invasive fungal disease. J Antimicrob Chemother 2017; 72: 3501. 10.1093/jac/dkx382 [DOI] [PubMed] [Google Scholar]
- 26. Furuno JP, Tallman GB, Noble BN et al. Clinical outcomes of oral suspension versus delayed-release tablet formulations of posaconazole for prophylaxis of invasive fungal infections. Antimicrob Agents Chemother 2018; 62: e00893-18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Tormo M, Pérez-Martínez A, Calabuig M et al. Primary prophylaxis of invasive fungal infections with posaconazole or itraconazole in patients with acute myeloid leukaemia or high-risk myelodysplastic syndromes undergoing intensive cytotoxic chemotherapy: a real-world comparison. Mycoses 2018; 61: 206–12. 10.1111/myc.12728 [DOI] [PubMed] [Google Scholar]
- 28. Yang E, Choi EJ, Park HS et al. Comparison of invasive fungal diseases between patients with acute myeloid leukemia receiving posaconazole prophylaxis and those not receiving prophylaxis: a single-center, observational, case-control study in South Korea. Medicine (Baltimore) 2021; 100: e25448. 10.1097/MD.0000000000025448 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. Fisher BT, Zaoutis T, Dvorak CC et al. Effect of caspofungin vs fluconazole prophylaxis on invasive fungal disease among children and young adults with acute myeloid leukemia: a randomized clinical trial. JAMA 2019; 322: 1673–81. 10.1001/jama.2019.15702 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Bogler Y, Stern A, Su Y et al. Efficacy and safety of isavuconazole compared with voriconazole as primary antifungal prophylaxis in allogeneic hematopoietic cell transplant recipients. Med Mycol 2021; 59: 970–9. 10.1093/mmy/myab025 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Stern A, Su Y, Lee YJ 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 Transplant 2020; 26: 1195–202. 10.1016/j.bbmt.2020.02.009 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Samanta P, Clancy CJ, Marini RV et al. Isavuconazole is as effective as and better tolerated than voriconazole for antifungal prophylaxis in lung transplant recipients. Clin Infect Dis 2021; 73: 416–26. 10.1093/cid/ciaa652 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Bose P, McCue D, Wurster S 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. 10.1093/cid/ciaa358 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Fontana L, Perlin DS, Zhao Y et al. Isavuconazole prophylaxis in patients with hematologic malignancies and hematopoietic cell transplant recipients. Clin Infect Dis 2020; 70: 723–30. 10.1093/cid/ciz282 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Cornely OA, Böhme A, Schmitt-Hoffmann A et al. Safety and pharmacokinetics of isavuconazole as antifungal prophylaxis in acute myeloid leukemia patients with neutropenia: results of a phase 2, dose escalation study. Antimicrob Agents Chemother 2015; 59: 2078–85. 10.1128/AAC.04569-14 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Hoenigl M, Salmanton-García J, Walsh TJ et al. Global guideline for the diagnosis and management of rare mould infections: an initiative of the European Confederation of Medical Mycology in cooperation with the International Society for Human and Animal Mycology and the American Society for Microbiology. Lancet Infect Dis 2021; 21: e246–e57. [DOI] [PubMed] [Google Scholar]
- 37. Ullmann AJ, Aguado JM, Arikan-Akdagli S et al. Diagnosis and management of Aspergillus diseases: executive summary of the 2017 ESCMID-ECMM-ERS guideline. Clin Microbiol Infect 2018; 24 Suppl 1: e1–e38. 10.1016/j.cmi.2018.01.002 [DOI] [PubMed] [Google Scholar]
- 38. Marks DI, Pagliuca A, Kibbler CC et al. Voriconazole versus itraconazole for antifungal prophylaxis following allogeneic haematopoietic stem-cell transplantation. Br J Haematol 2011; 155: 318–27. 10.1111/j.1365-2141.2011.08838.x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Bui V, Walker SA, Elligsen M et al. Voriconazole prophylaxis in leukemic patients: a retrospective single-center study. J Oncol Pharm Pract 2020; 26: 873–81. 10.1177/1078155219876683 [DOI] [PubMed] [Google Scholar]
- 40. Hachem R, Assaf A, Numan Y et al. Comparing the safety and efficacy of voriconazole versus posaconazole in the prevention of invasive fungal infections in high-risk patients with hematological malignancies. Int J Antimicrob Agents 2017; 50: 384–8. 10.1016/j.ijantimicag.2017.03.021 [DOI] [PubMed] [Google Scholar]
- 41. Xu XH, Zhang L, Cao XX et al. Evaluation of the implementation rate of primary antifungal prophylaxis and the prognosis of invasive fungal disease in acute leukemia patients in China. J Infect Chemother 2017; 23: 360–7. 10.1016/j.jiac.2017.02.011 [DOI] [PubMed] [Google Scholar]
- 42. Hicks JK, Quilitz RE, Komrokji RS et al. Prospective CYP2C19-guided voriconazole prophylaxis in patients with neutropenic acute myeloid leukemia reduces the incidence of subtherapeutic antifungal plasma concentrations. Clin Pharmacol Ther 2020; 107: 563–70. 10.1002/cpt.1641 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Tsutsumi I, Kunisawa S, Yoshida C et al. Impact of oral voriconazole during chemotherapy for acute myeloid leukemia and myelodysplastic syndrome: a Japanese nationwide retrospective cohort study. Int J Clin Oncol 2019; 24: 1449–58. 10.1007/s10147-019-01506-x [DOI] [PubMed] [Google Scholar]
- 44. Phillips K, Cirrone F, Ahuja T et al. Posaconazole versus voriconazole as antifungal prophylaxis during induction therapy for acute myelogenous leukemia or myelodysplastic syndrome. J Oncol Pharm Pract 2019; 25: 398–403. 10.1177/1078155218806975 [DOI] [PubMed] [Google Scholar]
- 45. Tang L, Yang XF, Qiao M et al. Posaconazole vs. voriconazole in the prevention of invasive fungal diseases in patients with haematological malignancies: a retrospective study. J Mycol Med 2018; 28: 379–83. 10.1016/j.mycmed.2017.11.003 [DOI] [PubMed] [Google Scholar]
- 46. Copley MS, Waldron M, Athans V 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. 10.1016/j.ijantimicag.2020.105886 [DOI] [PubMed] [Google Scholar]
- 47. Lindsay J, Sandaradura I, Wong K et al. Serum levels, safety and tolerability of new formulation SUBA-itraconazole prophylaxis in patients with haematological malignancy or undergoing allogeneic stem cell transplantation. J Antimicrob Chemother 2017; 72: 3414–9. 10.1093/jac/dkx295 [DOI] [PubMed] [Google Scholar]
- 48. 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. 10.1093/jac/dkz303 [DOI] [PubMed] [Google Scholar]
- 49. Whitmore TJ, Yaw M, Lavender M et al. A novel highly bio-available itraconazole formulation (SUBA®-itraconazole) for anti-fungal prophylaxis in lung transplant recipients. Transpl Infect Dis 2021; 23: e13587. 10.1111/tid.13587 [DOI] [PubMed] [Google Scholar]
- 50. Shen Loo Y, Yee Wong T, Veettil SK et al. Antifungal agents in preventing oral candidiasis in clinical oncology: a network meta-analysis. Oral Dis 2021; 27: 1631–43. 10.1111/odi.13588 [DOI] [PubMed] [Google Scholar]
- 51. Cornely FB, Cornely OA, Salmanton-García J et al. Attributable mortality of candidemia after introduction of echinocandins. Mycoses 2020; 63: 1373–81. 10.1111/myc.13177 [DOI] [PubMed] [Google Scholar]
- 52. Dvorak CC, Fisher BT, Esbenshade AJ et al. A randomized trial of caspofungin vs triazoles prophylaxis for invasive fungal disease in pediatric allogeneic hematopoietic cell transplant. J Pediatric Infect Dis Soc 2021; 10: 417–25. 10.1093/jpids/piaa119 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53. López-Sánchez C, Valcárcel D, Gómez V et al. Use of micafungin as antifungal prophylaxis in patients undergoing allogeneic hematopoietic stem cell transplantation (allo-HSCT) in Spain (GETH-MIC). Rev Esp Quimioter 2020; 33: 110–5. 10.37201/req/094.2019 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. Wingen-Heimann SM, Cornely OA, Vehreschild 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. 10.1111/myc.13232 [DOI] [PubMed] [Google Scholar]
- 55. Rothe A, Claßen A, Carney J et al. Bridging antifungal prophylaxis with 50 mg or 100 mg micafungin in allogeneic stem cell transplantation: a retrospective analysis. Eur J Haematol 2020; 104: 291–8. 10.1111/ejh.13372 [DOI] [PubMed] [Google Scholar]
- 56. Epstein DJ, Seo SK, Huang YT et al. Micafungin versus posaconazole prophylaxis in acute leukemia or myelodysplastic syndrome: a randomized study. J Infect 2018; 77: 227–34. 10.1016/j.jinf.2018.03.015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57. Kang WH, Song GW, Lee SG 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. 10.1007/s11605-019-04241-w [DOI] [PubMed] [Google Scholar]
- 58. Park S, Kim K, Jang JH et al. Randomized trial of micafungin versus fluconazole as prophylaxis against invasive fungal infections in hematopoietic stem cell transplant recipients. J Infect 2016; 73: 496–505. 10.1016/j.jinf.2016.06.011 [DOI] [PubMed] [Google Scholar]
- 59. Venton G, Adam H, Colle J et al. Micafungin as primary antifungal prophylaxis in patients presenting with acute myeloid leukemia. Med Mal Infect 2016; 46: 226–9. 10.1016/j.medmal.2016.03.008 [DOI] [PubMed] [Google Scholar]
- 60. Penack O, Schwartz S, Martus P et al. Low-dose liposomal amphotericin B in the prevention of invasive fungal infections in patients with prolonged neutropenia: results from a randomized, single-center trial. Ann Oncol 2006; 17: 1306–12. 10.1093/annonc/mdl128 [DOI] [PubMed] [Google Scholar]
- 61. Batchelor R, Thomas C, Gardiner BJ et al. When azoles cannot be used: the clinical effectiveness of intermittent liposomal amphotericin prophylaxis in hematology patients. Open Forum Infect Dis 2021; 8: ofab113. 10.1093/ofid/ofab113 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62. Luu Tran H, Mahmoudjafari Z, Rockey M et al. Tolerability and outcome of once weekly liposomal amphotericin B for the prevention of invasive fungal infections in hematopoietic stem cell transplant patients with graft-versus-host disease. J Oncol Pharm Pract 2016; 22: 228–34. 10.1177/1078155214560920 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63. Youngs J, Low JM, Whitney L et al. Safety and efficacy of intermittent high-dose liposomal amphotericin B antifungal prophylaxis in haemato-oncology: an eight-year single-centre experience and review of the literature. J Fungi (Basel) 2020; 6: 385. 10.3390/jof6040385 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64. Rijnders BJ, Cornelissen JJ, Slobbe L et al. Aerosolized liposomal amphotericin B for the prevention of invasive pulmonary aspergillosis during prolonged neutropenia: a randomized, placebo-controlled trial. Clin Infect Dis 2008; 46: 1401–8. 10.1086/586739 [DOI] [PubMed] [Google Scholar]
- 65. Chong GL, Broekman F, Polinder S et al. Aerosolised liposomal amphotericin B to prevent aspergillosis in acute myeloid leukaemia: efficacy and cost effectiveness in real-life. Int J Antimicrob Agents 2015; 46: 82–7. 10.1016/j.ijantimicag.2015.02.023 [DOI] [PubMed] [Google Scholar]
- 66. Gøtzsche PC, Johansen HK. Nystatin prophylaxis and treatment in severely immunodepressed patients. Cochrane Database Syst Rev 2014; 2014: Cd002033. [DOI] [PubMed] [Google Scholar]
- 67. Karthaus M, Lehrnbecher T, Lipp HP et al. Therapeutic drug monitoring in the treatment of invasive aspergillosis with voriconazole in cancer patients–an evidence-based approach. Ann Hematol 2015; 94: 547–56. 10.1007/s00277-015-2333-z [DOI] [PubMed] [Google Scholar]
- 68. Gómez-López A. Antifungal therapeutic drug monitoring: focus on drugs without a clear recommendation. Clin Microbiol Infect 2020; 26: 1481–7. 10.1016/j.cmi.2020.05.037 [DOI] [PubMed] [Google Scholar]
- 69. Kriegl L, Hatzl S, Zurl C et al. Isavuconazole plasma concentrations in critically ill patients during extracorporeal membrane oxygenation. J Antimicrob Chemother 2022; 77: 2500–5. 10.1093/jac/dkac196 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70. Prattes J, Duettmann W, Hoenigl M. Posaconazole plasma concentrations on days three to five predict steady-state levels. Antimicrob Agents Chemother 2016; 60: 5595–9. 10.1128/AAC.00389-16 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71. Stemler J, de Jonge N, Skoetz N et al. Antifungal prophylaxis in adult patients with acute myeloid leukaemia treated with novel targeted therapies: a systematic review and expert consensus recommendation from the European Hematology Association. Lancet Haematol 2022; 9: e361–e73. 10.1016/S2352-3026(22)00073-4 [DOI] [PubMed] [Google Scholar]
- 72. Agarwal SK, DiNardo CD, Potluri J et al. Management of venetoclax-posaconazole interaction in acute myeloid leukemia patients: evaluation of dose adjustments. Clin Ther 2017; 39: 359–67. 10.1016/j.clinthera.2017.01.003 [DOI] [PubMed] [Google Scholar]
- 73. Maiti A, Konopleva MY. How we incorporate venetoclax in treatment regimens for acute myeloid leukemia. Cancer J 2022; 28: 2–13. 10.1097/PPO.0000000000000567 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74. Rausch CR, DiNardo CD, Maiti A et al. Duration of cytopenias with concomitant venetoclax and azole antifungals in acute myeloid leukemia. Cancer 2021; 127: 2489–99. 10.1002/cncr.33508 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75. Rausch CR, Dinardo CD, Maiti A et al. Venetoclax dosing in combination with antifungal agents: real world experience in patients with acute myeloid leukemia. Blood 2019; 134 Suppl : 2640. 10.1182/blood-2019-131988 [DOI] [Google Scholar]
- 76. Sechaud R, Sinclair K, Grosch K et al. Evaluation of drug-drug interactions between midostaurin and strong CYP3A4 inhibitors in patients with FLT-3-mutated acute myeloid leukemia (AML). Cancer Chemother Pharmacol 2022; 90: 19–27. 10.1007/s00280-022-04448-w [DOI] [PubMed] [Google Scholar]
- 77. Aleissa MM, Alshehri BS, Gonzalez-Bocco IH et al. Triazole antifungal use for prophylaxis and treatment of invasive fungal diseases for patients receiving gilteritinib. Leuk Res 2021; 108: 106610. 10.1016/j.leukres.2021.106610 [DOI] [PubMed] [Google Scholar]
- 78. Child LA, Chan HSH, Henderson R et al. Single centre experience in treating elderly patients with acute myeloid leukaemia or high risk myelodysplastic syndrome with a combination of low dose cytarabine, venetoclax and fluconazole (VeLDAC-F). Blood 2018; 132 Suppl : 5220. 10.1182/blood-2018-99-115993 [DOI] [Google Scholar]
- 79. Cortes JE, Khaled S, Martinelli G et al. Quizartinib versus salvage chemotherapy in relapsed or refractory FLT3-ITD acute myeloid leukaemia (QuANTUM-R): a multicentre, randomised, controlled, open-label, phase 3 trial. Lancet Oncol 2019; 20: 984–97. 10.1016/S1470-2045(19)30150-0 [DOI] [PubMed] [Google Scholar]
- 80. Food and Drug Administration . Highlights of prescribing information - tibsovo (ivosidenib tablets), for oral use. 2021. Reference ID: 4847005.
- 81. Hoenigl M, Sprute R, Egger M et al. The antifungal pipeline: fosmanogepix, ibrexafungerp, olorofim, opelconazole, and rezafungin. Drugs 2021; 81: 1703–29. 10.1007/s40265-021-01611-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82. Cass L, Murray A, Davis A et al. Safety and nonclinical and clinical pharmacokinetics of PC945, a novel inhaled triazole antifungal agent. Pharmacol Res Perspect 2021; 9: e00690. 10.1002/prp2.690 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83. Thompson GR, Soriano A, Skoutelis A et al. Rezafungin versus caspofungin in a phase 2, randomized, double-blind study for the treatment of candidemia and invasive candidiasis: the STRIVE trial. Clin Infect Dis 2021; 73: e3647–e55. 10.1093/cid/ciaa1380 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84. Sonbol MB, Jain T, Firwana B et al. Neutropenic diets to prevent cancer infections: updated systematic review and meta-analysis. BMJ Support Palliat Care 2019; 9: 425–33. [DOI] [PubMed] [Google Scholar]
- 85. Gardner A, Mattiuzzi G, Faderl S et al. Randomized comparison of cooked and noncooked diets in patients undergoing remission induction therapy for acute myeloid leukemia. J Clin Oncol 2008; 26: 5684–8. 10.1200/JCO.2008.16.4681 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86. Jakob CEM, Classen AY, Stecher M et al. Association between the dietary regimen and infection-related complications in neutropenic high-risk patients with cancer. Eur J Cancer 2021; 155: 281–90. 10.1016/j.ejca.2021.06.054 [DOI] [PubMed] [Google Scholar]
- 87. Maschmeyer G, Neuburger S, Fritz L et al. A prospective, randomised study on the use of well-fitting masks for prevention of invasive aspergillosis in high-risk patients. Ann Oncol 2009; 20: 1560–4. 10.1093/annonc/mdp034 [DOI] [PubMed] [Google Scholar]
- 88. Combariza JF, Toro LF, Orozco JJ et al. Cost-effectiveness analysis of interventions for prevention of invasive aspergillosis among leukemia patients during hospital construction activities. Eur J Haematol 2018; 100: 140–6. 10.1111/ejh.12991 [DOI] [PubMed] [Google Scholar]
- 89. Passweg JR, Rowlings PA, Atkinson KA et al. Influence of protective isolation on outcome of allogeneic bone marrow transplantation for leukemia. Bone Marrow Transplant 1998; 21: 1231–8. 10.1038/sj.bmt.1701238 [DOI] [PubMed] [Google Scholar]
- 90. Özen M, Yılmaz G, Coşkun B et al. A quasi-experimental study analyzing the effectiveness of portable high-efficiency particulate absorption filters in preventing infections in hematology patients during construction. Turk J Haematol 2016; 33: 41–7. 10.4274/tjh.2014.0010 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91. Iwasaki M, Kanda J, Hishizawa M et al. Effect of laminar air flow and building construction on aspergillosis in acute leukemia patients: a retrospective cohort study. BMC Infect Dis 2019; 19: 38. 10.1186/s12879-018-3665-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92. Biehl LM, Huth A, Panse J et al. A randomized trial on chlorhexidine dressings for the prevention of catheter-related bloodstream infections in neutropenic patients. Ann Oncol 2016; 27: 1916–22. 10.1093/annonc/mdw275 [DOI] [PubMed] [Google Scholar]
- 93. Estcourt LJ, Stanworth S, Doree C et al. Granulocyte transfusions for preventing infections in people with neutropenia or neutrophil dysfunction. Cochrane Database Syst Rev 2015; 2015: Cd005341. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94. Desai PM, Brown J, Gill S et al. Open-label phase II prospective, randomized, controlled study of romyelocel-L myeloid progenitor cells to reduce infection during induction chemotherapy for acute myeloid leukemia. J Clin Oncol 2021; 39: 3261–72. 10.1200/JCO.20.01739 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95. Kang KW, Kim DS, Lee SR et al. Effect of granulocyte colony-stimulating factor on outcomes in patients with non-M3 acute myelogenous leukemia treated with anthracycline-based induction (7+3 regimen) chemotherapies. Leuk Res 2017; 57: 1–8. 10.1016/j.leukres.2017.02.003 [DOI] [PubMed] [Google Scholar]
- 96. Lien MY, Chou CH, Lin CC et al. Epidemiology and risk factors for invasive fungal infections during induction chemotherapy for newly diagnosed acute myeloid leukemia: a retrospective cohort study. PLoS ONE 2018; 13: e0197851. 10.1371/journal.pone.0197851 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97. Chien SH, Liu YC, Liu CJ et al. Invasive mold infections in acute leukemia patients undergoing allogeneic hematopoietic stem cell transplantation. J Microbiol Immunol Infect 2019; 52: 973–82. 10.1016/j.jmii.2018.09.006 [DOI] [PubMed] [Google Scholar]
- 98. Caira M, Candoni A, Verga L et al. Pre-chemotherapy risk factors for invasive fungal diseases: prospective analysis of 1,192 patients with newly diagnosed acute myeloid leukemia (SEIFEM 2010-a multicenter study). Haematologica 2015; 100: 284–92. 10.3324/haematol.2014.113399 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99. Lass-Flörl C, Dietl AM, Kontoyiannis DP et al. Aspergillus terreus species complex. Clin Microbiol Rev 2021; 34: e0031120. 10.1128/CMR.00311-20 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100. Bundesgesundheitsblatt Gesundheitsforschung Gesundheitsschutz 2021; 64: 232-64.
- 101. Combariza JF, Toro LF, Orozco JJ. Effectiveness of environmental control measures to decrease the risk of invasive aspergillosis in acute leukaemia patients during hospital building work. J Hosp Infect 2017; 96: 336–41. 10.1016/j.jhin.2017.04.022 [DOI] [PubMed] [Google Scholar]
- 102. Loschi M, Thill C, Gray C et al. Invasive aspergillosis in neutropenic patients during hospital renovation: effectiveness of mechanical preventive measures in a prospective cohort of 438 patients. Mycopathologia 2015; 179: 337–45. 10.1007/s11046-015-9865-6 [DOI] [PubMed] [Google Scholar]
- 103. Vehreschild JJ, Koehler P, Lamoth F et al. Future challenges and chances in the diagnosis and management of invasive mould infections in cancer patients. Med Mycol 2021; 59: 93–101. 10.1093/mmy/myaa079 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104. Glasmacher A, Prentice A, Gorschlüter 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: 4615–26. 10.1200/JCO.2003.04.052 [DOI] [PubMed] [Google Scholar]
- 105. Boogaerts MA, Verhoef GE, Zachee P et al. Antifungal prophylaxis with itraconazole in prolonged neutropenia: correlation with plasma levels. Mycoses 1989; 32 Suppl 1: 103–8. 10.1111/j.1439-0507.1989.tb02299.x [DOI] [PubMed] [Google Scholar]
- 106. Tricot G, Joosten E, Boogaerts MA et al. Ketoconazole vs. itraconazole for antifungal prophylaxis in patients with severe granulocytopenia: preliminary results of two nonrandomized studies. Rev Infect Dis 1987; 9 Suppl 1:S94–9. 10.1093/clinids/9.Supplement_1.S94 [DOI] [PubMed] [Google Scholar]
- 107. Schelenz S, Owens K, Guy R et al. National mycology laboratory diagnostic capacity for invasive fungal diseases in 2017: evidence of sub-optimal practice. J Infect 2019; 79: 167–73. 10.1016/j.jinf.2019.06.009 [DOI] [PubMed] [Google Scholar]
- 108. Risum M, Vestergaard MB, Weinreich UM et al. Therapeutic drug monitoring of isavuconazole: serum concentration variability and success rates for reaching target in comparison with voriconazole. Antibiotics (Basel) 2021; 10: 487. 10.3390/antibiotics10050487 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109. Czyrski A, Resztak M, Świderski P et al. The overview on the pharmacokinetic and pharmacodynamic interactions of triazoles. Pharmaceutics 2021; 13: 1961. 10.3390/pharmaceutics13111961 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110. Maertens JA, Raad II, Marr KA et al. Isavuconazole versus voriconazole for primary treatment of invasive mould disease caused by Aspergillus and other filamentous fungi (SECURE): a phase 3, randomised-controlled, non-inferiority trial. Lancet 2016; 387: 760–9. 10.1016/S0140-6736(15)01159-9 [DOI] [PubMed] [Google Scholar]
- 111. Andes D, Kovanda L, Desai A et al. Isavuconazole concentration in real-world practice: consistency with results from clinical trials. Antimicrob Agents Chemother 2018; 62: e00585-18. 10.1128/AAC.00585-18 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112. Desai A, Kovanda L, Kowalski D et al. Population pharmacokinetics of isavuconazole from phase 1 and phase 3 (SECURE) trials in adults and target attainment in patients with invasive infections due to Aspergillus and other filamentous fungi. Antimicrob Agents Chemother 2016; 60: 5483–91. 10.1128/AAC.02819-15 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113. Furfaro E, Signori A, Di Grazia C et al. Serial monitoring of isavuconazole blood levels during prolonged antifungal therapy. J Antimicrob Chemother 2019; 74: 2341–6. 10.1093/jac/dkz188 [DOI] [PubMed] [Google Scholar]
- 114. Tang LA, Marini BL, Benitez L et al. Risk factors for subtherapeutic levels of posaconazole tablet. J Antimicrob Chemother 2017; 72: 2902–5. 10.1093/jac/dkx228 [DOI] [PubMed] [Google Scholar]
- 115. Van Daele R, Spriet I, Maertens J. Posaconazole in prophylaxis and treatment of invasive fungal infections: a pharmacokinetic, pharmacodynamic and clinical evaluation. Expert Opin Drug Metab Toxicol 2020; 16: 539–50. 10.1080/17425255.2020.1764939 [DOI] [PubMed] [Google Scholar]
- 116. Ullmann AJ, Lipton JH, Vesole DH et al. Posaconazole or fluconazole for prophylaxis in severe graft-versus-host disease. N Engl J Med 2007; 356: 335–47. 10.1056/NEJMoa061098 [DOI] [PubMed] [Google Scholar]
- 117. Krishna G, Martinho M, Chandrasekar P et al. Pharmacokinetics of oral posaconazole in allogeneic hematopoietic stem cell transplant recipients with graft-versus-host disease. Pharmacotherapy 2007; 27: 1627–36. 10.1592/phco.27.12.1627 [DOI] [PubMed] [Google Scholar]
- 118. Jang SH, Colangelo PM, Gobburu JV. Exposure-response of posaconazole used for prophylaxis against invasive fungal infections: evaluating the need to adjust doses based on drug concentrations in plasma. Clin Pharmacol Ther 2010; 88: 115–9. 10.1038/clpt.2010.64 [DOI] [PubMed] [Google Scholar]
- 119. Chen L, Wang Y, Zhang T et al. Utility of posaconazole therapeutic drug monitoring and assessment of plasma concentration threshold for effective prophylaxis of invasive fungal infections: a meta-analysis with trial sequential analysis. BMC Infect Dis 2018; 18: 155. 10.1186/s12879-018-3055-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120. Märtson AG, Veringa A, van den Heuvel ER et al. Posaconazole therapeutic drug monitoring in clinical practice and longitudinal analysis of the effect of routine laboratory measurements on posaconazole concentrations. Mycoses 2019; 62: 698–705. 10.1111/myc.12948 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121. Suh HJ, Kim I, Cho JY et al. Early therapeutic drug monitoring of posaconazole oral suspension in patients with hematologic malignancies. Ther Drug Monit 2018; 40: 115–9. 10.1097/FTD.0000000000000469 [DOI] [PubMed] [Google Scholar]
- 122. Gautier-Veyret E, Bolcato L, Roustit M et al. Treatment by posaconazole tablets, compared to posaconazole suspension, does not reduce variability of posaconazole trough concentrations. Antimicrob Agents Chemother 2019; 63: e00484-19. 10.1128/AAC.00484-19 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123. Cornely OA, Helfgott D, Langston A et al. Pharmacokinetics of different dosing strategies of oral posaconazole in patients with compromised gastrointestinal function and who are at high risk for invasive fungal infection. Antimicrob Agents Chemother 2012; 56: 2652–8. 10.1128/AAC.05937-11 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124. Maleki S, Corallo C, Coutsouvelis J et al. Failure to achieve therapeutic levels with high-dose posaconazole tablets potentially due to enhanced clearance. J Oncol Pharm Pract 2018; 24: 63–6. 10.1177/1078155216673228 [DOI] [PubMed] [Google Scholar]
- 125. Conte JE Jr, Golden JA, Krishna G et al. Intrapulmonary pharmacokinetics and pharmacodynamics of posaconazole at steady state in healthy subjects. Antimicrob Agents Chemother 2009; 53: 703–7. 10.1128/AAC.00663-08 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126. Stott KE, Hope WW. Therapeutic drug monitoring for invasive mould infections and disease: pharmacokinetic and pharmacodynamic considerations. J Antimicrob Chemother 2017; 72: i12–i8. 10.1093/jac/dkx029 [DOI] [PubMed] [Google Scholar]
- 127. Wei X, Zhao M, Fu P et al. Risk factors associated with insufficient and potentially toxic voriconazole plasma concentrations: an observational study. J Chemother 2019; 31: 401–7. 10.1080/1120009X.2019.1646974 [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.