ArticlePDF Available

Ahead of consensus: A paediatric antifungal prophylaxis census

Authors:

Abstract

There is a strong argument for the use of antifungal prophylaxis in high-risk patients given the significant mortality associated with invasive fungal disease, the late identification of these infections, and the availability of safe and well-tolerated prophylactic medications. Clinical decisions about which patients should receive prophylaxis and choice of antifun-gal agent should be guided by risk stratification, knowledge of local fungal epidemiology, the efficacy and tolerability profile of available agents, and estimates such as number needed to treat and number needed to harm. There have been substantial changes in practice since the 2008 guidelines were published. These include the availability of new medications and/or formulations, and a focus on refining and simplifying patient risk stratification. Used in context, these guidelines aim to assist clinicians in providing optimal preventive care to this vulnerable patient demographic.
Ahead of consensus: a paediatric
antifungal prophylaxis census
We commend the authors of the Australian and New
Zealand Antifungal Consensus Guideline.1To context-
ualise this guideline in children, we surveyed current
routine primary antifungal prophylaxis practice for
patients with haematological malignancies at the seven
Australian children’s hospitals with major haematology-
oncology services across six states.
Formal guidelines were used at three of seven centres.
Conditions considered high risk for invasive fungal infec-
tion (IFI) were consistent with the consensus guideline,
with every hospital including acute myeloid leukaemia
(AML), myelodysplastic syndrome, aplastic anaemia,
high-risk acute lymphoblastic leukaemia (ALL), relapsed
leukaemia and post-haemopoietic stem cell transplant
(HSCT) with neutropenia or stage II-IV graft-versus-host
disease. Some hospitals identified other factors including
high-dose corticosteroids, prolonged neutropenia, young
age and comorbidities.
Prophylaxis type differed between conditions and hos-
pitals (Table 1). No hospital routinely offered prophylaxis
for patients with standard-risk ALL. One hospital used
yeast prophylaxis (i.e. fluconazole) with higher-intensity
chemotherapy regimens and another noted sporadic use
for patients perceived to be at higher risk (e.g. in
nappies). Of six institutions undertaking allogeneic HSCT,
three used yeast and three mould prophylaxis, including
one routinely using intermittent liposomal amphotericin.
Despite moderate evidence for decreased IFI in adult
autologous HSCT, yeast prophylaxis was used in only
three of six centres.2
Regarding choice of mould prophylaxis, five hospitals
preferentially used posaconazole for older children,
extrapolating from its success in adolescents and adults
with AML,3despite a lack of strong paediatric comparative
data.4Four of these hospitals used voriconazole for
younger patients and one itraconazole. Itraconazole was
used first-line for all ages at one institution. At another,
posaconazole was only used for older children with AML,
perhaps reflecting pharmaceutical benefit scheme restric-
tions. Two hospitals advised a cut-off age for posaconazole
use of >8 years and two used >13 years, while noting some
use under these age cut-offs, reflecting the still-limited
paediatric pharmacokinetic data available.5–7 For high-risk
patients not tolerating oral intake, voriconazole, liposomal
amphotericin and caspofungin were used. For patients
receiving vinca alkaloids, most hospitals use intermittent
liposomal amphotericin despite lack of strong evidence for
this strategy.8–11
Therapeutic drug monitoring (TDM) was readily acces-
sible for azoles at all centres. Two hospitals used serum
galactomannan for screening (for some groups) and diag-
nosis, one hospital planned to do so and three other
hospitals only used galactomannan as a diagnostic test.
One hospital did not use it. TDM and galactomannan
turnaround times ranged from 24 h to 5 days and 48 h to
7 days, respectively, with wide variations even within
institutions, depending on onsite availability and labora-
tory schedules.
Prophylaxis can be an effective strategy for reduc-
ing the impact of IFI for vulnerable paediatric
haematology-oncology patients. Areas of uncertainty
include risk stratification, choice of mould prophylaxis,
practical application of TDM and biomarkers,
posaconazole pharmacokinetics, and use of non-azole
agents. The consensus guideline provides a uniform
approach based largely on expert opinion due to scarce
paediatric data. The need remains for prospective data
collection in children. Study of prevention and manage-
ment of infection should be integrated into compr-
ehensive paediatric haematology-oncology research
protocols.
Table 1 Spectrum of routine primary antifungal prophylaxis by condition and hospital
Hospitals
AB C D E F G
ALL – standard risk None None None/yeast† None None None None/yeast†
ALL – high risk Mould None None/yeast† Mould None/yeast/mould† Mould Yeast/mould†
AML Mould Mould None/mould† Mould Yeast Mould Mould
Relapsed leukaemia Mould Mould Mould Mould Mould Mould Mould
AA/MDS Mould Mould Yeast Mould Mould Mould Mould
Autologous HSCT‡ None N/a None None Yeast Yeast Yeast
Allogeneic HSCT‡ Yeast N/a Mould Yeast Yeast/mould† Mould Yeast
†Dependent on patient (e.g. Down syndrome, children in nappies), intensity of chemotherapy and protocol factors. ‡Neutropenic phase. AA, aplastic
anaemia; ALL, acute lymphoblastic leukaemia; AML, acute myeloid leukaemia; HSCT,haemopoietic stem cell transplant ; MDS, myelodysplastic syndrome.
bs_bs_banner
Letters to the Editor
© 2015 Royal Australasian College of Physicians
364
Received 9 December 2014; accepted 21 December 2014.
doi:10.1111/imj.12690
J. Osowicki,1,2 C. C. Blyth,3,4,5,6 P. N. Britton,7,8 J. Clark,9
C. M. Cooper,10 G. M. Haeusler,11,12 B. McMullan13,14
and P. A. Bryant1,2,15
1Infectious Diseases Unit, The Royal Children’s Hospital
Melbourne, 2Murdoch Children’s Research Institute, 15Department
of Paediatrics, The University of Melbourne, 11Department of
Paediatric Infectious Diseases, Monash Children’s Hospital,
12Department of Infectious Diseases, Peter MacCallum Cancer
Centre, Melbourne, Victoria, 3Department of General Paediatrics,
Princess Margaret Hospital for Children, 5PathWest Laboratory
Medicine WA, Princess Margaret Hospital, Perth, 4School of
Paediatrics and Child Health, University of Western Australia,
6Telethon Kids Institute, University of Western Australia, , Perth,
Western Australia, 7Department of Infectious Diseases and
Microbiology, The Children’s Hospital at Sydney, Westmead,
8Discipline of Paediatrics and Child Health, Sydney Medical School,
University of Sydney, 14School of Women’s and Children’s Health,
University of New South Wales, 13Department of Immunology and
Infectious Diseases, Sydney Children’s Hospital, Sydney, New
South Wales, 9Infection Management and Prevention Service, Lady
Cilento Children’s Hospital, Brisbane, Queensland, and 10SA
Pathology, Women’s and Children’s Hospital, Adelaide, South
Australia, Australia
References
1 Slavin MA, Thursky KA, Worth LJ,
Chang CC, Morrissey CO, Blyth CC et al.
Revised Australian and New Zealand
antifungal consensus guideline 2014.
Intern Med J 2014; 44: 1267–397.
2 Goodman JL, Winston DJ, Greenfield
RA, Chandrasekar PH, Fox B, Kaizer H
et al. A controlled trial of fluconazole to
prevent fungal infections in patients
undergoing bone marrow
transplantation. N Engl J Med 1992; 326:
845–51.
3 Cornely OA, Maertens J, Winston DJ,
Perfect J, Ullmann AJ, Walsh TJ et al.
Posaconazole vs. fluconazole or
itraconazole prophylaxis in patients with
neutropenia. N Engl J Med 2007; 356:
348–59.
4 Doring M, Blume O, Haufe S, Hartmann
U, Kimmig A, Schwarze CP et al.
Comparison of itraconazole,
voriconazole, and posaconazole as oral
antifungal prophylaxis in pediatric
patients following allogeneic
hematopoietic stem cell transplantation.
Eur J Clin Microbiol Infect Dis 2014; 33:
629–38.
5 Doring M, Muller C, Johann PD,
Erbacher A, Kimmig A, Schwarze CP
et al. Analysis of posaconazole as oral
antifungal prophylaxis in pediatric
patients under 12 years of age following
allogeneic stem cell transplantation. BMC
Infect Dis 2012; 12: 263.
6 Krishna G, Sansone-Parsons A,
Martinho M, Kantesaria B, Pedicone L.
Posaconazole plasma concentrations in
juvenile patients with invasive fungal
infection. Antimicrob Agents Chemother
2007; 51: 812–18.
7 Bernardo VA, Cross SJ, Crews KR, Flynn
PM, Hoffman JM, Knapp KM et al.
Posaconazole therapeutic drug
monitoring in pediatric patients and
young adults with cancer. Ann
Pharmacother 2013; 47: 976–83.
8 Moriyama B, Henning SA, Leung J,
Falade-Nwulia O, Jarosinski P, Penzak
SR et al. Adverse interactions between
antifungal azoles and vincristine: review
and analysis of cases. Mycoses 2012; 55:
290–7.
9 Bochennek K, Tramsen L, Schedler N,
Becker M, Klingebiel T, Groll AH et al.
Liposomal amphotericin B twice weekly
as antifungal prophylaxis in paediatric
haematological malignancy patients.
Clini Microbiol Infect 2011; 17: 1868–74.
10 Roman E, Osunkwo I, Militano O,
Cooney E, van de Ven C, Cairo MS.
Liposomal amphotericin B prophylaxis
of invasive mold infections in children
post allogeneic stem cell trans-
plantation. Pediatr Blood Cancer 2008; 50:
325–30.
11 Uhlenbrock S, Zimmermann M, Fegeler
W, Jurgens H, Ritter J. Liposomal
amphotericin B for prophylaxis of
invasive fungal infections in high-risk
paediatric patients with
chemotherapy-related neutropenia:
interim analysis of a prospective study.
Mycoses 2001; 44: 455–63.
Regional differences in
ventilation-perfusion ratio may help
explain the differential diagnosis in
interstitial lung disease
Interstitial lung disease is a well-recognised respiratory
condition often seen in respiratory and general medicine
clinics. Not surprisingly, because their physical signs on
chest examination are relatively stable, and due to the fact
that their investigations offer a multitude of potential
questions relating to diagnosis and management, patients
with interstitial lung disease are often invited to partici-
pate in the Fellowship of the Royal Australasian College of
Physicians (clinical examination, part 1). Over the past 25
years, a whole generation of doctors, who grew up with
the highly regarded textbooks written by Nicholas Talley
and Simon O’Connor,1,2 have used the mnemonics
SCHART (for upper lobe predominant) and RASIO (for
lower lobe-predominant interstitial lung disease) to aid
prompt recitation of the differential diagnosis of interstitial
lung disease (Table 1). The utility of all mnemonics,
however, is affected by time, due to changes in disease
prevalence. Pneumoconioses, in particular, have become
much less common in the past decade,3especially with
improvements in mining technology and disease preven-
tion efforts. Epidemiology data relating to the prevalence
bs_bs_banner
Letters to the Editor
© 2015 Royal Australasian College of Physicians
365
Article
Invasive fungal disease (IFD) remains a common and serious complication in children treated for leukaemia. Antifungal prescription in children with leukaemia presents unique challenges, particularly due to variation in IFD risk between and within leukaemia treatment protocols, drug toxicities and interactions between antifungals and chemotherapeutic agents. With recent advances in the understanding of IFD epidemiology and large clinical trials in adults assessing antifungals for IFD treatment and prophylaxis, together with paediatric clinical and pharmacokinetic studies, there is a growing body of data to inform optimal antifungal use in children. A panel of infectious diseases and haematology-oncology clinicians with expertise in IFD management compiled a list of 10 key clinical questions following development of the 2021 Australia and New Zealand Mycology Antifungal Consensus Guidelines. A focused literature review was conducted to explore available evidence and identify gaps in knowledge to direct future research. With the changing epidemiology of IFD globally, the ongoing evolution of paediatric leukaemia treatment and the increasing availability of novel antifungal agents, advocacy for paediatric clinical studies will remain vital to optimize IFD prevention and treatment in children with leukaemia.
Article
Background Invasive fungal disease (IFD) is a common and important complication in children with acute myeloid leukaemia (AML). We describe the epidemiology of IFD in a large multicentre cohort of children with AML. Methods As part of the retrospective multicentre cohort TERIFIC (The Epidemiology and Risk factors for Invasive Fungal Infections in immunocompromised Children) study, proven/probable/possible IFD episodes occurring in children with primary or relapsed/refractory AML from 2003 to 2014 were analysed. Crude IFD prevalence, clinical characteristics, microbiology and treatment were assessed. Kaplan–Meier survival analysis was used to estimate 6-month survival. Results There were 66 IFD episodes diagnosed in 63 children with AML. The majority (75.8%) of episodes occurred in the context of primary AML therapy. During primary AML therapy, the overall prevalence was 20.7% (95% CI 15.7%–26.5%) for proven/probable/possible IFD and 10.3% (95% CI 6.7%–15.0%) for proven/probable IFD. Of primary AML patients, 8.2% had IFD diagnosed during the first cycle of chemotherapy. Amongst pathogens implicated in proven/probable IFD episodes, 74.4% were moulds, over a third (37.9%) of which were non-Aspergillus spp. Antifungal prophylaxis preceded 89.4% of IFD episodes, most commonly using fluconazole (50% of IFD episodes). All-cause mortality at 6 months from IFD diagnosis was 16.7% with IFD-related mortality of 7.6% (all in cases of proven IFD). Conclusions IFD is a common and serious complication during paediatric AML therapy. Mould infections, including non-Aspergillus spp. predominated in this cohort. A systematic approach to the identification of patients at risk, and a targeted prevention strategy for IFD is needed.
Article
Full-text available
Oral antifungal prophylaxis with extended-spectra azoles is widely used in pediatric patients after allogeneic hematopoietic stem cell transplantation (HSCT), while controlled studies for oral antifungal prophylaxis after bone marrow transplantation in children are not available. This survey analyzed patients who had received either itraconazole, voriconazole, or posaconazole. We focused on the safety, feasibility, and initial data of efficacy in a cohort of pediatric patients and adolescents after high-dose chemotherapy and HSCT. Fifty consecutive pediatric patients received itraconazole, 50 received voriconazole, and 50 pediatric patients received posaconazole after HSCT as oral antifungal prophylaxis. The observation period lasted from the start of oral prophylactic treatment with itraconazole, voriconazole, or posaconazole until two weeks after terminating the oral antifungal prophylaxis. No incidences of proven or probable invasive mycosis were observed during itraconazole, voriconazole, or posaconazole treatment. A total of five possible invasive fungal infections occurred, two in the itraconazole group (4 %) and three in the voriconazole group (6 %). The percentage of patients with adverse events potentially related to clinical drugs were 14 % in the voriconazole group, 12 % in the itraconazole group, and 8 % in the posaconazole group. Itraconazole, voriconazole, and posaconazole showed comparable efficacy as antifungal prophylaxis in pediatric patients after allogeneic HSCT.
Article
Full-text available
BACKGROUND Limited information exists regarding the use of posaconazole for treating systemic fungal infections in children, adolescents, and young adults with cancer. At St. Jude Children's Research Hospital, the recommended posaconazole dose in patients weighing less than 34 kg is 18–24 mg/kg daily, given in 4 divided doses. For patients aged 13 years or older or those weighing 34 kg or more, the recommended dose is 800 mg daily, given orally in 4 divided doses. OBJECTIVE To determine whether the current posaconazole dosing guidelines achieve target posaconazole plasma concentrations of 0.7 μg/mL or greater. METHODS This retrospective clinical study examined data from patients who received treatment-dose posaconazole and had at least 1 posaconazole plasma concentration measurement. RESULTS Data from 33 patients who received posaconazole for the treatment of fungal infections were analyzed. The median age of patients was 11.5 years (range 0.5–23.2). Twenty-one of 33 patients (63.6%) had posaconazole concentrations of 0.7 μg/mL or greater (median 1.4; range 0.7–2.98) at the first measurement. The median posaconazole dosage referenced to total body weight in these patients was 20 mg/kg/day. Patients with concentrations less than 0.7 μg/mL (median 0.4; range 0.025–0.69) received lower posaconazole dosages when referenced to body weight (median 12.9 mg/kg/day; p = 0.02). Of the 12 patients with concentrations less than 0.7 μg/mL, 7 (58.3%) were aged 13 years or older. CONCLUSIONS The current dosing approach for posaconazole yielded therapeutic plasma concentrations more frequently in patients younger than 13 years than in those 13 years or older. This difference may be related to the practice of capping adolescent and young adult doses at the suggested maximum adult daily dose. Therefore, we recommend weight-based dosing in all pediatric, adolescent, and young adult patients with cancer, with routine therapeutic drug monitoring to ensure adequate concentrations.
Article
Full-text available
Background Pediatric patients undergoing hematopoietic stem cell transplantation (HSCT) are at high risk of acquiring fungal infections. Antifungal prophylaxis shortly after transplantation is therefore indicated, but data for pediatric patients under 12 years of age are scarce. To address this issue, we retrospectively assessed the safety, feasibility, and initial efficacy of prophylactic posaconazole in children. Methods 60 consecutive pediatric patients with a median age of 6.0 years who underwent allogeneic HSCT between August 2007 and July 2010 received antifungal prophylaxis with posaconazole in the outpatient setting. 28 pediatric patients received an oral suspension at 5 mg/kg body weight b.i.d., and 32 pediatric patients received the suspension at 4 mg/kg body weight t.i.d. The observation period lasted from start of treatment with posaconazole until its termination (maximum of 200 days post-transplant). Results Pediatric patients who received posaconazole at 4 mg/kg body weight t.i.d. had a median trough level of 383 μg/L. Patients who received posaconazole at 5 mg/kg body weight b.i.d. had a median trough level of 134 μg/L. Both regimens were well tolerated without severe side effects. In addition, no proven or probable invasive mycosis was observed. Conclusion Posaconazole was a well-tolerated, safe, and effective oral antifungal prophylaxis in pediatric patients who underwent high-dose chemotherapy and HSCT. Posaconazole at a dosage of 12 mg/kg body weight divided in three doses produced consistently higher morning trough levels than in patients who received posaconazole 5 mg/kg body weight b.i.d. Larger prospective trials are needed to obtain reliable guidelines for antifungal prophylaxis in children after HSCT.
Article
Full-text available
Clin Microbiol Infect 2011; 17: 1868–1874 Data on antifungal prophylaxis in paediatric cancer patients at high risk for invasive fungal disease (IFD) are scant. Intermittent administration of liposomal amphotericin B (LAMB) has been shown to be safe and effective in adult patients with haematological malignancies. We prospectively evaluated the safety and efficacy of prophylactic LAMB at a dosage of 2.5 mg/kg twice weekly in children at high risk for IFD. Efficacy was compared with that in a historical control group of patients with similar demographic characteristics not receiving LAMB prophylaxis. A total of 46 high-risk patients (24 boys; mean age, 7.7 years) with 187 episodes of antifungal prophylaxis were analysed. The median duration of neutropenia (<500/μL) was 10 days. LAMB was discontinued in four patients because of acute allergic reactions. Median values for creatinine and liver enzymes at end of treatment did not differ significantly from those at baseline. Hypokalaemia (<3.0 mmol/L) occurred with 13.5% of the prophylactic episodes, but was usually mild and always reversible. No proven/probable IFD occurred in patients receiving LAMB prophylaxis. In comparison, five proven and two probable IFDs were observed in 45 historical controls not receiving LAMB prophylaxis (p 0.01). LAMB prophylaxis had no impact on the use of empirical antifungal therapy. Systemic antifungal prophylaxis with LAMB 2.5 mg/kg twice weekly is feasible and safe, and seems to be an effective approach for antifungal prophylaxis in high-risk paediatric cancer patients.
Article
Full-text available
Superficial and systemic fungal infections are a major problem among severely immunocompromised patients who undergo bone marrow transplantation. We performed a double-blind, randomized, multicenter trial in which patients receiving bone marrow transplants were randomly assigned to receive placebo or fluconazole (400 mg daily). Fluconazole or placebo was administered prophylactically from the start of the conditioning regimen until the neutrophil count returned to 1000 per microliter, toxicity was suspected, or a systemic fungal infection was suspected or proved. By the end of the treatment period, 67.2 percent of the 177 patients assigned to placebo had a positive fungal culture of specimens from any site, as compared with 29.6 percent of the 179 patients assigned to fluconazole. Among these, superficial infections were diagnosed in 33.3 percent of the patients receiving placebo and in 8.4 percent of the patients receiving fluconazole (P less than 0.001). Systemic fungal infections occurred in 28 patients who received placebo as compared with 5 who received fluconazole (15.8 percent vs. 2.8 percent, P less than 0.001). Fluconazole prevented infection with all strains of candida except Candida krusei. Fluconazole was well tolerated, although patients who received it had a higher mean increase in alanine aminotransferase levels than patients who received placebo. Although there was no significant difference in overall mortality between the groups, fewer deaths were ascribed to acute systemic fungal infections in the group receiving fluconazole than in the group receiving placebo (1 of 179 vs. 10 of 177, P less than 0.001). Prophylactic administration of fluconazole to recipients of bone marrow transplants reduces the incidence of both systemic and superficial fungal infections.
Article
This article introduces the second revision of the Australian and New Zealand consensus guidelines for the use of antifungal agents in the haematology/oncology setting. The current update occurs within the context of a growing population at risk of invasive fungal disease, improved understanding of risk factors, availability of new diagnostic tests, a much-expanded evidence base and changing clinical paradigms. Here, we provide an overview of the history and purpose of the guidelines, including changes in scope since the last clinical update was published in 2008. The process for development, and for enabling review of draft recommendations by end-users and other relevant stakeholders, is described. The approach to assigning levels of evidence and grades of recommendation is also provided, along with a comparison to international grading systems. © 2014 The Authors; Internal Medicine Journal © 2014 Royal Australasian College of Physicians.
Article
Invasive fungal infections (IFIs) are a major cause of morbidity and mortality in paediatric acute myeloid leukaemia (AML). This study describes risk factors for IFI and IFI-related sepsis in this population. We conducted a population-based, retrospective cohort study of children with AML in Canada. IFIs during chemotherapy and prior to haematopoietic stem cell transplantation, relapse, persistent disease or death were identified. Risk factors for proven or probable IFI were examined. Among courses complicated by IFI, risk factors for sepsis were also evaluated. There were 341 children with AML included of which 41 (12.0%) experienced 46 different episodes of IFI. Candida species accounted for 23 (50.0%) of IFIs and Aspergillus spp. accounted for 14 (30.4%). Days of broad-spectrum antibiotics, days of corticosteroids and neutropenia at start of the course were independently associated with IFI. Only days of fever were independently associated with IFI-related sepsis. Invasive fungal infections occurred in 12.0% of paediatric AML patients. Risk factors for IFI and IFI-related sepsis were identified. This knowledge may help to consider targeted strategies.
Article
Background Invasive mold infections (IMI) are a leading cause of infectious mortality in allogeneic stem cell transplant (AlloSCT) recipients. Fluconazole, the current standard for fungal prophylaxis, is ineffective against molds. We initiated a pilot study to determine the safety and activity of prophylactic liposomal amphotericin B (AMB) in preventing IMI in pediatric and adolescent AlloSCT recipients during the first 100 days.ProcedureFifty-one patients (57 AlloSCT) were given AMB (3 mg/kg/day) intravenously, day 0–100. Median age 6 years, 32 males, 19 females. Donors: 33 unrelated and 2 related cord blood, 13 related and 1 unrelated peripheral blood stem cell and 8 related bone marrow (BM); 30 received myeloablative and 27 reduced intensity conditioning. Graft-versus-host disease (GVHD) prophylaxis comprised tacrolimus and mycophenolate mofetil.ResultsMedian follow-up is 557 days. AMB was generally well tolerated. The probability of developing ≥grade II acute GVHD and extensive chronic GVHD was 45% and 7%, respectively. Estimated 1-year OS is 62.4% for all patients with 78.8% and 26.7% for average-risk and poor-risk, respectively. The incidence of IMI was 0%.Conclusions These results suggest prophylactic AMB is tolerable and may prevent IMI, especially Aspergillus, during the first 100 days post AlloSCT in pediatric and adolescent patients. A randomized study is needed to determine the efficacy of this approach. Pediatr Blood Cancer 2008;50:325–330. © 2007 Wiley-Liss, Inc.
Article
Triazole and imidazole antifungal agents inhibit metabolism of vincristine, leading to excess vinca alkaloid exposure and severe neurotoxicity. Recent reports of debilitating interactions between vincristine and itraconazole, as well as posaconazole, voriconazole and ketoconazole underscore the need to improve medical awareness of this adverse combination. We, therefore, undertook a comprehensive analysis of reports of adverse drug interactions (ADIs) with the combination of vincristine and azole antifungal agents, established a new classification, and provided a detailed summary of these toxicities. In patients who had sufficient data for analysis, 47 individuals were identified who had an ADI with the combination of vincristine and antifungal azoles. Median age was 8 years (1.3-68 years) with 33(70%) having a diagnosis of acute lymphoblastic leukaemia. Median time to ADI with vincristine was 9.5 days with itraconazole, 13.5 days posaconazole and 30 days voriconazole. The median number of vincristine doses preceding the ADI was 2 doses with itraconazole, 3 doses posaconazole and 2 doses voriconazole. The most common severe ADIs included gastrointestinal toxicity, peripheral neuropathy, hyponatremia/SIADH, autonomic neuropathy and seizures. Recovery from these ADIs occurred in 80.6% of patients. We recommend using alternative antifungal agents if possible in patients receiving vincristine to avoid this serious and potentially life-threatening drug interaction.
Article
The epidemiology, management, and long-term survival of invasive aspergillosis was assessed in a prospective, 5-year observational study in 346 unselected paediatric cancer patients receiving dose-intensive chemotherapy for newly diagnosed or recurrent malignancies. Invasive aspergillosis occurred exclusively in the context of haematological malignancies, where it accounted for an incidence of 6.8% (n = 13 of 189). The lung was the primary site in 12 cases, and dissemination was present in three of those. Prior to diagnosis, the overwhelming majority of patients had been profoundly neutropenic for at least 14 days (n = 11 of 13) and were receiving systemic antifungal agents (n = 10 of 13). Clinical signs and symptoms were nonspecific but always included fever. All 11 patients who were diagnosed and treated during lifetime for a minimum of 10 days responded to either medical or combined medical and surgical treatment, and seven were cured (64%). Nevertheless, the overall long-term survival was merely 31% after a median follow-up of 5.68 years after diagnosis. Apart from refractory or recurrent cancer, the main obstacles to successful outcome were failure to diagnose IA during lifetime and bleeding complications in patients with established diagnosis. The frequency of invasive aspergillosis of greater than 15% in paediatric patients with acute myeloblastic leukaemia and recurrent leukaemias warrants the systematic investigation of preventive strategies in these highly vulnerable subgroups.