Skip to main content

Clinical application of voriconazole in pediatric patients: a systematic review

Abstract

The purpose of this study was to review the literature on the clinical use of voriconazole (VRC) in pediatric patients. MEDLINE, Embase, PubMed, Web of Science, and Cochrane Library were searched from January 1, 2000, to August 15, 2023 for relevant clinical studies on VRC use in pediatric patients. Data were collected based on inclusion and exclusion criteria, and a systematic review was performed on recent research related to the use of VRC in pediatric patients. This systematic review included a total of 35 observational studies among which there were 16 studies investigating factors influencing VRC plasma trough concentrations (Ctrough) in pediatric patients, 14 studies exploring VRC maintenance doses required to achieve target range of Ctrough, and 11 studies focusing on population pharmacokinetic (PPK) research of VRC in pediatric patients. Our study found that the Ctrough of VRC were influenced by both genetic and non-genetic factors. The optimal dosing of VRC was correlated with age in pediatric patients, and younger children usually required higher VRC doses to achieve target Ctrough compared to older children. Establishing a PPK model for VRC can assist in achieving more precise individualized dosing in children.

Introduction

Voriconazole (VRC) is a broad-spectrum triazole antifungal agent, primarily used for the treatment of progressive and potentially life-threatening fungal infections, as well as for the prevention of invasive fungal infections (IFIs) in high-risk patients undergoing allogeneic hematopoietic stem cell transplantation (allo-HSCT) [1, 2]. The Infectious Diseases Society of America (IDSA) and the European Conference on Infections in Leukaemia (ECIL) all recommended VRC as the preferred treatment for invasive aspergillosis (IA) [3, 4].

VRC is rapidly and completely absorbed through oral administration, and it is widely distributed in tissues. However, due to its narrow therapeutic window and significant inter- and intra-individual variability in plasma trough concentrations (Ctrough) [5], personalized dosing strategies should be implemented to ensure efficacy and reduce adverse reactions. In recent years, there have been numerous studies related to VRC therapeutic drug monitoring (TDM), population pharmacokinetics (PPK) analysis and pharmacogenomics in children. Research on the factors affecting VRC Ctrough and dose optimization has been constantly being updated.

As a special population in terms of medication, ensuring the safety and efficacy of VRC use is of utmost importance in pediatric patients. The VRC use in pediatric patients has gained the increasing attention of researchers. There were currently many studies on VRC use in children. However, without summarizing these findings, clinicians or pharmacists may lack sufficient understanding of the characteristics of VRC use in pediatric patients, potentially hindering the achievement of personalized dosing.

Therefore, we need to summarize the research on pediatric VRC use. The aim of this review was to provide guidance for improving the effectiveness and safety of VRC in pediatric patients and to establish a theoretical basis for achieving personalized dosing in clinical therapeutics.

Methods

The authors identified three key questions:

  1. i.

    What dosage is required to attain the target Ctrough of VRC?

  2. ii.

    What factors influence VRC Ctrough in pediatric patients?

  3. iii.

    What recommendations can be derived from the PPK study of VRC for personalized medication?

Search strategy

We conducted a systematic review in accordance with the guidelines outlined in the Preferred Reporting Items for Systematic Reviews and Meta-analyses (PRISMA) [6]. We conducted computer searches in databases, such as MEDLINE, EMbase, PubMed, Web of science and Cochrane Library databases, with a search period spanning from January 1, 2000, to August 15, 2023. Duplicate articles found in different databases were removed by Using EndNote. Based on the characteristics of different databases, corresponding search strategies were formulated to preliminarily screen literature related to the use of VRC in pediatric patients. The search terms were as follows: (voriconazole) AND (children) OR (child) OR (pediatric patient) OR (infant) OR (adolescent) AND (factor) OR (influence) OR (affect) OR (effect) OR (population pharmacokinetic) OR (PPK) OR (dose optimization) OR (dosage optimization).

Study selection

All articles describing factors influencing VRC Ctrough, dose optimization and PPK studies were included in this review. The inclusion criteria: (1) the study drug must be VRC, and steady-state Ctrough must be monitored. (2) the study population must contain patients aged 0 to 18 years. (3) articles must be written in English. The exclusion criteria: (1) in vitro and animal studies. (2) reviews, systematic reviews, meta-analyses, letters, comments or case reports.

Data extraction

According to the purpose and specific content of this review, a uniform data extraction table was formulated. Two authors recorded the following information of included studies: authors, publication dates, countries, study design, sample sizes, patient characteristics such as underlying diseases and range of age, target range of VRC steady-state Ctrough, factors significantly influencing VRC Ctrough, VRC dosages, administration routes, durations of VRC use, software and models used in PPK studies, significant covariates affecting pharmacokinetic (PK) parameters and main results or conclusions of dose simulation experiments. Any disputed issues were discussed and resolved by the third author. We would not conduct further statistical analysis of the research data mentioned in this review and the results were displayed in tables.

Quality evaluation of studies

Observational studies were evaluated for adherence to the Strengthening the Reporting of Observational Studies in Epidemiology (STROBE) guidelines [7].

Results

Study selection

A total of 3 669 relevant articles were searched from the database (120 from MEDLINE, 2 240 from Embase, 956 from PubMed, 669 from Web of Science, and 43 from the Cochrane Library). According to the criteria of inclusion and exclusion, a total of 35 observational studies [8,9,10,11,12,13,14,15,16,17,18,19,20,21,22,23,24,25,26,27,28,29,30,31,32,33,34,35,36,37,38,39,40,41,42] remained in the systematic review after excluding 3 634 articles. The process and outcomes of literature screening were presented in Fig. 1, while the quality assessment of the selected studies was reported in Fig. 2.

Fig. 1
figure 1

The flowchart of articles selection

Fig. 2
figure 2

Adherence to STROBE recommendations

What dosage is required to attain the target Ctrough of VRC?

The maintenance doses of VRC required to achieve target range of Ctrough in both Asian (7 published studies) and non-Asian (7 published studies) pediatric patients were significantly correlated with age, as detailed in Tables 1 and 2. Boast et al. [20] found that due to the higher clearance rate (CL) and larger apparent volume of distribution in younger children compared to older children, the median intravenous dosages required to achieve target Ctrough for Australia patients aged < 6, 6–12 and > 12 years were 8.8, 7.5 and 4.0 mg/kg twice daily, respectiveiy (P < 0.001). Bartelink et al. [18] discovered that the average dosages required to achieve target Ctrough in Dutch children aged < 2, 2–12 and > 12 years were 31.5, 16.0 and 9.4 mg/kg/day, respectively, with statistically significant differences in daily dosages among the three age groups. Similar results were observed in our previous studies involving Chinese pediatric patients [11, 14]. The above researches all found that younger children required higher doses to achieve the target VRC Ctrough compared to older children. Therefore, both Asian and non-Asian pediatric patients required individualized VRC dosing regimens based on age.

Table 1 Summary of studies on VRC maintenance doses to achieve the target range in Asian populations
Table 2 Summary of studies on VRC maintenance doses to achieve the target range in non-Asian populations

We have also observed that the maintenance doses of VRC required to achieve target Ctrough might differ between Asian and non-Asian pediatric patients. A retrospective study from China found that intravenous dosages of 5–7 mg/kg twice daily could satisfy the requirements for achieving target Ctrough in most Asian pediatric patients [10]. Hu et al. [11] also discovered that the oral and intravenous dosages needed to achieve target Ctrough in pediatric patients were significantly lower than the recommended dosages in European or American package inserts (7.7 mg/kg vs. 9 mg/kg, P = 0.033; 5.6 mg/kg vs. 8 mg/kg, P = 0.003). However, due to the unavailability of data for further statistical analysis, it remains uncertain whether differences exist in the VRC doses needed to achieve target Ctrough between Asian and non-Asian pediatric populations.

What factors influence VRC Ctrough in pediatric patients?

The Ctrough of VRC were influenced by various factors in pediatric patients. Currently, there have been 16 published studies investigating the determinants of VRC Ctrough in pediatric patients, among which only 2 were prospective studies, while the rest were retrospective, single-center and descriptive studies. Those studies came from various regions: Asia (n = 11; 68.8%), encompassing 9 from China, 1 from Japan, and 1 from Korea; Europe (n = 4; 25.0%), comprising two in Italy and one each in Spain and Switzerland; and South America (n = 1; 6.2%), specifically from Chile. Six studies had included sample sizes of over 100 pediatric patients, and merely two studies had encompassed sample sizes exceeding 200 pediatric patients. These investigations have identified more than ten factors that could significantly impact VRC Ctrough, as outlined in Table 3. The most frequently reported significant influencing factors including CYP2C19 genetic polymorphism, co-administration of proton pump inhibitors (PPIs), inflammation status and liver function indicators.

Table 3 Summary of studies exploring the factors affecting the VRC Ctrough in pediatric patients

Genetic factors significantly influenced the metabolism of VRC. Numerous studies have shown a significant correlation between CYP2C19 genetic polymorphism and VRC Ctrough in pediatric patients. Studies by Espinoza et al. [24] and Fan et al. [31] have found that mutations such as CYP2C19*2 and CYP2C19*3 might lead to decreased enzyme activity and increased VRC Ctrough, while the CYP2C19*17 mutation might result in enhanced enzyme activity and decreased VRC Ctrough. Chen et al.‘s study [29] found that 15.3% of patients were CYP2C19 poor metabolizers (PMs), a proportion higher than that reported in European and American populations. Allegra et al. [23] and Tilen et al. [30] reported that apart from CYP2C19 genotypes, genetic polymorphisms in CYP3A4, SLCO1B3, as well as ABCC2 and ABCG2 also significantly influenced VRC Ctrough.

A retrospective single-center study conducted in 2017 involving Chinese pediatric patients aged 0–12 years demonstrated that concurrent administration of omeprazole significantly elevated VRC Ctrough (P = 0.032), providing the evidence of the impact of omeprazole on VRC Ctrough in pediatric patients [10]. Hu et al. [11] found that concomitant use of PPIs significantly elevated Ctrough of VRC (median VRC Ctrough in patients with and without PPIs co-administration were 2.07 mg/L vs. 0.84 mg/L, respectively, P = 0.028) through a retrospective analysis. Co-administration of VRC and PPIs lead to a significant increase in VRC Ctrough.

Currently, four studies have reported the correlation between C-reactive protein (CRP) concentrations and VRC Ctrough in pediatric patients. A clinical study found differences in the correlation between the CRP concentrations and VRC Ctrough among pediatric patients of different age groups. Luo et al. [28] discovered a significant correlation between CRP concentrations and VRC PK in pediatric patients aged 11–18 years, but no significant correlation was observed in patients aged 2–10 years.

Due to the nonlinear PK of VRC, Ctrough could not be predicted by dose. Moreover, most studies indicated that VRC dosage was unrelated to Ctrough. However, the CL of VRC may exhibit linearity in the pediatric population. Liu et al. [10] discovered that no correlation between VRC Ctrough and dose in pediatric patients aged 2–12 years (n = 27, r = 0.151, P = 0.452), however, a notable correlation was observed between VRC Ctrough and dosage (n = 74, r = 0.370, P = 0.001) in pediatric patients < 2 years old. Allegra et al. [22] also found a significant correlation between VRC Ctrough and dosage in pediatric patients (n = 237, r = 0.195, P = 0.016) in pediatric patients < 18 years old. Hence, the PK of VRC in pediatric patients may differ from those in adults.

VRC Ctrough were related to the routes of administration. Several studies have explored the impact of administration routes on VRC Ctrough. Patients receiving intravenous administration exhibited significantly higher VRC Ctrough compared to those receiving oral administration. Research by Allegra et al. demonstrated a positive correlation between VRC Ctrough and age in 237 Italian pediatric patients [22]. Furthermore, VRC Ctrough may also be associated with gender, liver and kidney function indicators. Allegra et al. also found that VRC Ctrough were significantly higher in males compared to females [22], while Liu et al. confirmed a significant positive correlation between VRC Ctrough and serum creatinine (Scr), and a significant negative correlation with serum albumin (ALB) levels [10]. Kang et al. [25] found a significant positive correlation between VRC Ctrough and aspartate aminotransferase (AST) levels.

What recommendations can be derived from the PPK study of VRC for personalized medication?

Currently, a total of 11 studies have established PPK models for pediatric patients. Nine studies used a two-compartment model and the most commonly used tool in PPK studies was non-linear mixed effect modeling (NONMEM). Among the 11 studies, 8 studies utilized NONMEM and only one PPK model incorporated CRP concentrations into covariance analysis [41], as detailed in Table 4. High inter-individual variability in VRC PK among the pediatric population had been revealed. Most of studies have identified CYP2C19 genetic polymorphisms as significant covariates influencing the PK parameters of VRC [32, 33, 35, 39, 40, 42]. Furthermore, covariates including body weight, age, CRP concentrations, co-administration of omeprazole, and liver function indicators such as ALB, alanine transaminase (ALT), and alkaline phosphatase (ALP) levels, may also be associated with VRC PK parameters [32,33,34,35,36, 39,40,41,42].

Table 4 Summary of studies on PPK analysis and current recommendations of VRC optimal dosing regimen in pediatric patients

Some studies employed the final models to explore optimal dosing regimens through dose simulation experiments. For instance, studies by Takahashi et al. [39], Wang et al. [40], and Wu et al. [42] proposed dose recommendations based on body weight and CYP2C19 genetic polymorphisms. All three studies recommended lower VRC doses for CYP2C19 PMs. Karlsson et al. [33] and Gastine et al. [37] directly provided simple and unified dose recommendations. Studies by Walsh et al. [32] and Friberg et al. [35] suggested dosing regimens in pediatric patients to achieve VRC exposures comparable to those in adults. Moreover, some studies proposed dose optimization suggestions based on other significant covariates. Wang et al. [40] suggested a slight reduction in VRC dose when co-administered with omeprazole, while Wu et al. [42] proposed that children with lower body weight might require higher VRC doses and those with low ALB levels might need lower VRC doses. By comparing estimated PK parameters between adults and pediatric patients, we found that PK parameters in children might differ from those in adults. Muto et al.‘s study [36] investigated the metabolic characteristics of VRC in Japanese pediatric immunocompromised patients, revealing an average bioavailability of 73% in this group, whereas it was 96% in healthy adult patients. Gastine et al.‘s study [37] estimated an average bioavailability of 59.4%. However, Wu et al.‘s study [42], which focused on the Chinese pediatric population, demonstrated that the bioavailability in pediatric patients could reach 90.2%.

Discussion

At present, research concerning the utilization of VRC in pediatric patients is garnering heightened attention. Investigations into the factors influencing VRC Ctrough, along with PPK analyses, serve as pivotal guides for dose optimization. Nonetheless, the realm of VRC utilization in pediatric patients with challenges like limited sample sizes and a preponderance of retrospective studies. These hurdles underscore the necessity for further comprehensive exploration within this special population.

i. What dosage is required to attain the target Ctrough of VRC?

Differences in VRC dosing exist between Asian and non-Asian pediatric patients, which may be attributed to variations in genetic backgrounds between these populations. Since VRC was predominantly metabolized by the liver enzyme CYP2C19, the proportion of Asians with the CYP2C19 PMs ranged from 15 to 20%, whereas in Caucasians, it was 3–5% [43]. This divergence could lead to differences in VRC metabolism among different ethnicities and subsequently resulted in variations in the required dosages to achieve target Ctrough. Asian pediatric patients may not be suited for the recommended dosages stated in the original manufacturer’s instructions.

The latest consensus by the JSC/JSTDM (2022) [44] suggested the necessity of reducing the standard dose for Asian populations due to the observed high incidence of supertherapeutic concentrations in TDM practice in Japan. Moreover, the consensus emphasized the need for distinct dosing regimens tailored to Asian and non-Asian populations to prevent overdosing. In the future, it is hoped that large-scale, cross-ethnicity prospective studies will be conducted to explore optimal dosages of VRC for diverse pediatric populations worldwide.

In addition, studies have indicated that pediatric patients needed to be administered appropriate dosages based on their age. Younger children may exhibit higher CL of VRC compared to older children, potentially necessitating different VRC doses among age groups. Nevertheless, guidelines have yet to specify reference VRC doses for pediatric patients (< 6, 6–12, > 12 years old). Furthermore, according to the FDA drug label information [45], it was important to consider that pediatric patients may have shorter gastrointestinal transit times, possibly affecting tablet absorption compared to adults. As a result, oral suspension was recommended for pediatric patients aged 2 to 12 years. However, the bioequivalence or PK studies between oral tablets and suspension of VRC has not been investigated in pediatric populations.

ii. What factors influence VRC Ctrough in pediatric patients?

When assessing factors influencing VRC Ctrough, although most of the studies were retrospective and single-center, they confirmed the already well-known factors such as CYP2C19 polymorphisms, concurrent use of PPIs, and patient age. Additionally, new factors including other genetic polymorphisms, CRP concentration, liver and kidney function, as well as gender, have been identified.

Weiss et al. proposed that CYP2C19 genotype significantly contributed to the high variability observed in VRC PK [46]. Trubiano et al. [47] also suggested that the CYP2C19 genotype could be utilized to predict VRC Ctrough and toxicity. Many studies suggested using CYP2C19 genotype to guide the initial dosing regimen of VRC [48, 49]. A study involving prophylactic use of VRC in acute myeloid leukemia patients found that CYP2C19 genotype testing not only avoided prolonging hospital stays but also moderately reduced costs, and it was projected that each patient could save $ 415 in hospitalization expenses [49].

The variability of VRC Ctrough can not be fully explained by concomitant medications, genetic polymorphisms of metabolic enzyme, or liver disorders. Recent researches indicated a correlation between elevated CRP concentration and lower VRC Ctrough. Morgan et al. [50] suggested that the release of cytokines upon inflammatory stimulation altered the activity of transcription factors in the liver. These alterations lead to the downregulation of most CYP genes, affecting the production of metabolic proteins and subsequently reducing the CL of VRC. In vitro studies have provided compelling evidence indicating that pro-inflammatory cytokines, especially interleukin-1 (IL-1), IL-6, and tumor necrosis factor-alpha (TNF-α), downregulated the biosynthesis of CYP isoforms, including CYP2C19, CYP3A4, and CYP2C9, which play pivotal roles in VRC metabolism [51, 52]. The correlation between CRP concentrations and VRC Ctrough showed variations in different age groups of pediatric patients. This discrepancy may be attributed to the distinct roles of CYP2C19, CYP3A4, and flavin-containing monooxygenase 3 (FMO-3) in VRC N-oxidation between pediatric patients and adults. Studies have found that the CL of VRC in patients aged 2 to 11 years was nearly three times that of adults [47]. CYP2C19 and FMO-3 exhibited higher metabolic activity in young children, and the downregulation of CYP2C19 isoforms during inflammation had a relatively minor impact on VRC metabolism in younger children. Further research is needed to explore how to achieve personalized dosing of VRC based on inflammatory status.

Although CYP2C19 enzymes accounted for only 5% of drug metabolism [46], they were involved in the metabolism of various drugs such as PPIs, antiepileptic drugs, antiplatelet drugs, and antidepressants. PPIs and corticosteroids being the most studied drugs that interact with VRC. The guideline issued by the Chinese Pharmacological Society (CPS) recommended closely monitoring the efficacy and safety of VRC when administered concomitantly with PPIs or corticosteroids [53].

The VRC Ctrough in pediatric patients were correlated with indicators of hepatic and renal function, indicating that elevated VRC Ctrough might be linked to impaired hepatic and renal function. For pediatric patients with normal renal function, the drug label recommended intravenous treatment for at least the initial 7 days of therapy for those with IA. Subsequently, upon clinical improvement and tolerance of oral medication, the oral tablet or suspension forms of VRC may be utilized. However, injectable VRC with the solvent sulfobutylether-β-cyclodextrin has been associated with adverse effects on kidney function due to potential accumulation. Research conducted by Yasu et al. [54] has demonstrated a significant correlation between renal function deterioration and cumulative intravenous VRC dose (≥ 400 mg/kg). These findings indicated that higher cumulative intravenous VRC doses may contribute to the risk of impaired kidney function. The FDA drug instructions advised careful attention was required when administering VRC intravenous preparations to patients with renal insufficiency (creatinine clearance rate < 50 ml/min) [45]. However, the long-term effects of intravenous VRC use on kidney function remain unclear. Currently, there is limited research on the use of VRC in pediatric patients with impaired hepatic or renal function.

iii. What recommendations can be derived from the PPK study or guidelines of VRC for personalized medication?

Despite the high inter-individual PK variability of VRC, PPK software for individualized dosing can accurately simulate VRC Ctrough, with predicted levels closely aligning with actual measured values. PPK model may be an immensely useful tool for further optimizing VRC dosing and assisting in TDM for clinical therapies. Further prospective research is required to determine its role in clinical practice. Utilizing the PPK model to describe patients’ PK characteristics and examining covariates significantly influencing VRC Ctrough can provide essential information for formulating individualized dosing regimens. The guideline of CPS recommended adjusting VRC dosing based on a PPK model for the Chinese population [53]. Therefore, PPK analysis for VRC in children is an important direction in future research.

Numerous PPK studies have emphasized CYP2C19 polymorphism as a significant covariate influencing the PK parameters of VRC, and some have proposed dosing regimens based on different CYP2C19 genotypes through dose simulation experiments. However, determining the initial dose by detecting CYP2C19 genotype is not yet recommended in the FDA drug label. The Clinical Pharmacogenetics Implementation Consortium (CPIC) guideline [55] provided dosing optimization schemes for VRC treatment based on CYP2C19 phenotype in patients aged < 18 years. For CYP2C19 rapid metabolizers (RMs), normal metabolizers (NMs), and intermediate metabolizers (IMs), initiating treatment with standard doses was recommended, with TDM advised for RMs to adjust the dose to achieve therapeutic Ctrough. In cases where VRC use was unavoidable for PMs, a reduced standard dose and TDM were recommended. Ultra-rapid metabolizers (UMs) were advised to switch to alternative drugs that did not undergo CYP2C19 metabolism, such as amphotericin B and posaconazole.

Previous research had proposed that the CL of VRC in pediatric patients was three times that of adults [56]. Studies by Pascual et al. [57] and Wang et al. [58] on adult patients reported VRC CL of 5.2 L/h and 6.95 L/h, respectively. However, research by Takahashi et al. estimated a VRC CL of 12.3 L/h for pediatric patients. Numerous PPK studies also suggested that pediatric patients often require higher doses than adults in order to achieve the same VRC exposure. In the 2013 Guideline for Japan [59], children were recommended to receive a dosage of 7 mg/kg q12h, which was lower than the dosage specified in the FDA drug label information. However, both the ESCMID-ECMM [60] and UK [61] guidelines advocated a loading dose of 9 mg/kg q12h, followed by a maintenance dose of 8 mg/kg q12h for the intravenous preparation, with oral dosing maintained at 9 mg/kg q12h, consistent with the dosage stated in the original manufacturer’s instructions. The latest consensus suggested that altering the initial VRC dose when coadministered with PPIs might be unnecessary until the results of TDM were available. The impact of CRP levels on the VRC Ctrough has been confirmed in numerous studies. However, many PPK studies of VRC did not include CRP concentrations. Hence, future PPK studies should consider incorporating inflammatory indicators such as CRP concentrations.

Conclusions

In recent years, due to the widespread of TDM and CYP2C19 genotype testing for VRC, the realization of VRC personalized therapies has become a prominent research focus. VRC Ctrough exhibit high inter- and intra-individual variability, potentially influenced by various factors such as age, concomitant medications, inflammatory status, hepatic and renal functions, as well as genetic polymorphisms in metabolic enzyme. Some unknown influencing factors need to be explored in the further studies. It is anticipated that more studies on personalized therapy of VRC will emerge, contributing to a comprehensive understanding of the factors influencing VRC Ctrough and PK variability.

Data availability

Not applicable.

References

  1. Walsh TJ, Anaissie EJ, Denning DW, Herbrecht R, Kontoyiannis DP, Marr KA, Morrison VA, Segal BH, Steinbach WJ, Stevens DA, van Burik JA, Wingard JR, Patterson TF. Infectious Diseases Society of America. Treatment of aspergillosis: clinical practice guidelines of the Infectious Diseases Society of America. Clin Infect Dis. 2008;46(3):327–60.

    Article  CAS  PubMed  Google Scholar 

  2. Trifilio SM, Yarnold PR, Scheetz MH, Pi J, Pennick G, Mehta J. Serial plasma voriconazole concentrations after allogeneic hematopoietic stem cell transplantation. Antimicrob Agents Chemother. 2009;53(5):1793–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Patterson TF, Thompson GR 3rd, Denning DW, Fishman JA, Hadley S, Herbrecht R, Kontoyiannis DP, Marr KA, Morrison VA, Nguyen MH, Segal BH, Steinbach WJ, Stevens DA, Walsh TJ, Wingard JR, Young JA, Bennett JE. Practice guidelines for the diagnosis and management of aspergillosis: 2016 update by the Infectious Diseases Society of America. Clin Infect Dis. 2016;63(4):e1–60.

    Article  PubMed  PubMed Central  Google Scholar 

  4. Groll AH, Castagnola E, Cesaro S, Dalle JH, Engelhard D, Hope W, Roilides E, Styczynski J, Warris A, Lehrnbecher T, Fourth European Conference on Infections in Leukaemia; Infectious Diseases Working Party of the European Group for Blood Marrow Transplantation (EBMT-IDWP); Infectious Diseases Group of the European Organisation for Research and Treatment of Cancer (EORTC-IDG). ; International Immunocompromised Host Society (ICHS); European Leukaemia Net (ELN). Fourth European Conference on Infections in Leukaemia (ECIL-4): guidelines for diagnosis, prevention, and treatment of invasive fungal diseases in paediatric patients with cancer or allogeneic haemopoietic stem-cell transplantation. Lancet Oncol. 2014; 15(8):e327-40.

  5. Dolton MJ, Mikus G, Weiss J, Ray JE, McLachlan AJ. Understanding variability with voriconazole using a population pharmacokinetic approach: implications for optimal dosing. J Antimicrob Chemother. 2014;69(6):1633–41.

    Article  CAS  PubMed  Google Scholar 

  6. Page MJ, McKenzie JE, Bossuyt PM, Boutron I, Hoffmann TC, Mulrow CD, Shamseer L, Tetzlaff JM, Akl EA, Brennan SE, Chou R, Glanville J, Grimshaw JM, Hróbjartsson A, Lalu MM, Li T, Loder EW, Mayo-Wilson E, McDonald S, McGuinness LA, Stewart LA, Thomas J, Tricco AC, Welch VA, Whiting P, Moher D. The PRISMA 2020 statement: an updated guideline for reporting systematic reviews. BMJ. 2021;372:n71.

    Article  PubMed  PubMed Central  Google Scholar 

  7. Adams AD, Benner RS, Riggs TW, Chescheir NC. Use of the STROBE Checklist to evaluate the Reporting Quality of Observational Research in Obstetrics. Obstet Gynecol. 2018;132(2):507–12.

    Article  PubMed  Google Scholar 

  8. Choi SH, Lee SY, Hwang JY, Lee SH, Yoo KH, Sung KW, Koo HH, Kim YJ. Importance of voriconazole therapeutic drug monitoring in pediatric cancer patients with invasive aspergillosis. Pediatr Blood Cancer. 2013;60(1):82–7.

    Article  CAS  PubMed  Google Scholar 

  9. Kato K, Nagao M, Yamamoto M, Matsumura Y, Takakura S, Fukuda K, Ichiyama S. Oral administration and younger age decrease plasma concentrations of voriconazole in pediatric patients. J Infect Chemother. 2016;22(1):27–31.

    Article  CAS  PubMed  Google Scholar 

  10. Liu L, Zhou X, Wu T, Jiang H, Yang S, Zhang Y. Dose optimisation of voriconazole with therapeutic drug monitoring in children: a single-centre experience in China. Int J Antimicrob Agents. 2017;49(4):483–7.

    Article  CAS  PubMed  Google Scholar 

  11. Hu L, Dai T-T, Zou L, Li T-M, Ding X-S, Yin T. Therapeutic drug monitoring of voriconazole in children from a tertiary care center in China. Antimicrob Agents Chemother. 2018;62:e00955–18.

    Article  PubMed  PubMed Central  Google Scholar 

  12. Tian X, Zhang C, Qin Z, Wang D, Yang J, Zhang X. Impact of CYP2C19 phenotype and drug-drug interactions on Voriconazole Concentration in Pediatric patients. Antimicrob Agents Chemother. 2021;65(9):e0020721.

    Article  PubMed  Google Scholar 

  13. Chen J, Wu Y, He Y, Feng X, Ren Y, Liu S. Combined effect of CYP2C19 genetic polymorphisms and C-Reactive protein on Voriconazole exposure and dosing in Immunocompromised Children. Front Pediatr. 2022;10:846411.

    Article  PubMed  PubMed Central  Google Scholar 

  14. Hu L, Huang Q, Huang S, Feng Z. Therapeutic drug monitoring of voriconazole and CYP2C19 phenotype for dose optimization in paediatric patients. Eur J Clin Pharmacol. 2023;79(9):1271–8.

    Article  CAS  PubMed  Google Scholar 

  15. Gerin M, Mahlaoui N, Elie C, Lanternier F, Bougnoux ME, Blanche S, Lortholary O, Jullien V. Therapeutic drug monitoring of voriconazole after intravenous administration in infants and children with primary immunodeficiency. Ther Drug Monit. 2011;33(4):464–6.

    Article  CAS  PubMed  Google Scholar 

  16. Soler-Palacín P, Frick MA, Martín-Nalda A, Lanaspa M, Pou L, Roselló E, de Heredia CD, Figueras C. Voriconazole drug monitoring in the management of invasive fungal infection in immunocompromised children: a prospective study. J Antimicrob Chemother. 2012;67(3):700–6.

    Article  PubMed  Google Scholar 

  17. Pieper S, Kolve H, Gumbinger HG, Goletz G, Würthwein G, Groll AH. Monitoring of voriconazole plasma concentrations in immunocompromised paediatric patients. J Antimicrob Chemother. 2012;67(11):2717–24.

    Article  CAS  PubMed  Google Scholar 

  18. Bartelink IH, Wolfs T, Jonker M, de Waal M, Egberts TC, Ververs TT, Boelens JJ, Bierings M. Highly variable plasma concentrations of voriconazole in pediatric hematopoietic stem cell transplantation patients. Antimicrob Agents Chemother. 2013;57(1):235–40.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Tucker L, Higgins T, Egelund EF, Zou B, Vijayan V, Peloquin CA. Voriconazole monitoring in children with invasive fungal infections. J Pediatr Pharmacol Ther. 2015;20(1):17–23.

    PubMed  PubMed Central  Google Scholar 

  20. Boast A, Curtis N, Cranswick N, Gwee A. Voriconazole dosing and therapeutic drug monitoring in children: experience from a paediatric tertiary care centre. J Antimicrob Chemother. 2016;71(7):2031–6.

    Article  CAS  PubMed  Google Scholar 

  21. Zembles TN, Dasgupta M, Kenkel TJ, Lehrer B, Simpson P, Havens PL, Huppler AR. Higher weight-based doses are required to Achieve and maintain therapeutic voriconazole serum trough concentrations in children. J Pediatr Pharmacol Ther. 2023;28(3):247–54.

    PubMed  PubMed Central  Google Scholar 

  22. Allegra S, Fatiguso G, De Francia S, Favata F, Pirro E, Carcieri C, De Nicolò A, Cusato J, Di Perri G, D’Avolio A. Therapeutic drug monitoring of voriconazole for treatment and prophylaxis of invasive fungal infection in children. Br J Clin Pharmacol. 2018;84(1):197–203.

    Article  CAS  PubMed  Google Scholar 

  23. Allegra S, Fatiguso G, Francia S, Pirro E, Carcieri C, Cusato J, Nicolò A, Avataneo V, Perri GD, D’Avolio A. Pharmacogenetic of voriconazole antifungal agent in pediatric patients. Pharmacogenomics. 2018;19(11):913–25.

    Article  CAS  PubMed  Google Scholar 

  24. Espinoza N, Galdames J, Navea D, Farfán MJ, Salas C. Frequency of the CYP2C19*17 polymorphism in a Chilean population and its effect on voriconazole plasma concentration in immunocompromised children. Sci Rep. 2019;9(1):8863.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Kang S, Yee J, Kim JY, Han HW, Kang SO, Lee KE, Gwak HS. Factors Associated with Voriconazole Concentration in Pediatric patients. Ther Drug Monit. 2020;42(6):866–71.

    Article  CAS  PubMed  Google Scholar 

  26. Zhao YC, Zou Y, Hou JJ, Xiao CL, Zhang BK, Li JK, Xiang DX, Sandaradura I, Yan M. Factors affecting voriconazole trough concentration and optimal maintenance voriconazole dose in Chinese children. Antibiot (Basel). 2021;10(12):1542.

    Article  CAS  Google Scholar 

  27. Valle-T-Figueras JM, Renedo Miró B, Benítez Carabante MI, Díaz-de-Heredia C, Vima Bofarull J, Mendoza-Palomar N, Martín-Gómez MT. Soler-Palacín P. Voriconazole Use in Children: therapeutic drug monitoring and Control of Inflammation as key points for optimal treatment. J Fungi (Basel). 2021;7(6):456.

    Article  CAS  PubMed  Google Scholar 

  28. Luo X, Li T, Hu L, Liu S, Zhao H, Zhang J, Feng Y, Huang L. Differential effects of C-reactive protein levels on voriconazole metabolism at three age groups in allogeneic hematopoietic cell transplant recipients. J Chemother. 2021;33(2):95–105.

    Article  CAS  PubMed  Google Scholar 

  29. Chen X, Xiao Y, Li H, Huang Z, Gao J, Zhang X, Li Y, Van Timothee BM, Feng X. Therapeutic drug monitoring and CYP2C19 genotyping guide the application of voriconazole in children. Transl Pediatr. 2022;11(8):1311–22.

    Article  PubMed  PubMed Central  Google Scholar 

  30. Tilen R, Paioni P, Goetschi AN, Goers R, Seibert I, Müller D, Bielicki JA, Berger C, Krämer SD. Meyer Zu Schwabedissen HE. Pharmacogenetic Analysis of Voriconazole Treatment in Children. Pharmaceutics. 2022;14(6):1289.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Fan X, Zhang H, Wen Z, Zheng X, Yang Y, Yang J. Effects of CYP2C19, CYP2C9 and CYP3A4 gene polymorphisms on plasma voriconazole levels in Chinese pediatric patients. Pharmacogenet Genomics. 2022;32(4):152–8.

    Article  CAS  PubMed  Google Scholar 

  32. Walsh TJ, Karlsson MO, Driscoll T, Arguedas AG, Adamson P, Saez-Llorens X, Vora AJ, Arrieta AC, Blumer J, Lutsar I, Milligan P, Wood N. Pharmacokinetics and safety of intravenous voriconazole in children after single- or multiple-dose administration. Antimicrob Agents Chemother. 2004;48(6):2166–72.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Karlsson MO, Lutsar I, Milligan PA. Population pharmacokinetic analysis of voriconazole plasma concentration data from pediatric studies. Antimicrob Agents Chemother. 2009;53(3):935–44.

    Article  CAS  PubMed  Google Scholar 

  34. Neely M, Rushing T, Kovacs A, Jelliffe R, Hoffman J. Voriconazole pharmacokinetics and pharmacodynamics in children. Clin Infect Dis. 2010;50(1):27–36.

    Article  CAS  PubMed  Google Scholar 

  35. Friberg LE, Ravva P, Karlsson MO, Liu P. Integrated population pharmacokinetic analysis of voriconazole in children, adolescents, and adults. Antimicrob Agents Chemother. 2012;56(6):3032–42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Muto C, Shoji S, Tomono Y, Liu P. Population pharmacokinetic analysis of voriconazole from a pharmacokinetic study with immunocompromised Japanese pediatric subjects. Antimicrob Agents Chemother. 2015;59:3216–23.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Gastine S, Lehrnbecher T, Müller C, Farowski F, Bader P, Ullmann-Moskovits J, Cornely OA, Groll AH, Hempel G. Pharmacokinetic modeling of voriconazole to develop an alternative dosing regimen in children. Antimicrob Agents Chemother. 2018;62:e01194–17.

    Article  PubMed  Google Scholar 

  38. Carlesse FAMC, de Araujo OR, Marques LMA, Silva DCBD, Senerchia AA, Petrilli AS. A pharmacokinetic model for voriconazole in a highly diversified population of children and adolescents with cancer. Mycoses. 2019;62(4):399–404.

    Article  CAS  PubMed  Google Scholar 

  39. Takahashi T, Mohamud MA, Smith AR, Jacobson PA, Jaber MM, Alharbi AF, Fisher J, Kirstein MN. CYP2C19 phenotype and body weight-guided Voriconazole initial dose in infants and children after hematopoietic cell transplantation. Antimicrob Agents Chemother. 2021;65(9):e0062321.

    Article  PubMed  Google Scholar 

  40. Wang J, Xu H, Li R, Wu S, Zou J. WangY. Model-oriented dose optimization of voriconazole in critically ill children. Antimicrob Agents Chemother. 2021;65:e00493–21.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Takahashi T, Jaber MM, Smith AR, Jacobson PA, Fisher J, Kirstein MN. Predictive value of C-Reactive protein and albumin for temporal within-individual pharmacokinetic variability of VRC in Pediatric patients undergoing hematopoietic cell transplantation. J Clin Pharmacol. 2022;62(7):855–62.

    Article  CAS  PubMed  Google Scholar 

  42. Wu Y, Lv C, Wu D, Qi J, Cai R, Zhou S, Li C, Wei Y, Liu T. Dosage optimization of voriconazole in children with haematological malignancies based on population pharmacokinetics. J Clin Pharm Ther. 2022; 47(12):2245–2254.

  43. PharmGKB.org. Gene-specific information tables for CYP2C19 [online]. Available at http://www.pharmgkb.org/page/cyp2c19RefMaterials (accessed 4 January 2019).

  44. Takesue Y, Hanai Y, Oda K, Hamada Y, Ueda T, Mayumi T, Matsumoto K, Fujii S, Takahashi Y, Miyazaki Y, Kimura T, Japanese Antimicrobial Therapeutic Drug Monitoring Guideline Committee. Clinical practice Guideline for the Therapeutic Drug Monitoring of Voriconazole in non-asian and Asian adult patients: Consensus Review by the Japanese Society of Chemotherapy and the Japanese Society of Therapeutic Drug Monitoring. Clin Ther. 2022;44(12):1604–23.

    Article  CAS  PubMed  Google Scholar 

  45. FDA drug label information of voriconazole. https://dailymed.nlm.nih.gov/dailymed/drugInfo.cfm?setid=63473087-5cc1-b2e8-b552-6d5f733e1227 [Accessed August 15, 2023].

  46. Weiss J, Ten Hoevel MM, Burhenne J, Walter-Sack I, Hoffmann MM, Rengelshausen J, Haefeli WE, Mikus G. CYP2C19 genotype is a major factor contributing to the highly variable pharmacokinetics of voriconazole. J Clin Pharmacol. 2009;49(2):196–204.

    Article  CAS  PubMed  Google Scholar 

  47. Trubiano JA, Crowe A, Worth LJ, Thursky KA, Slavin MA. Putting CYP2C19 genotyping to the test: utility of pharmacogenomic evaluation in a voriconazole-treated haematology cohort. J Antimicrob Chemother. 2015;70(4):1161–5.

    Article  CAS  PubMed  Google Scholar 

  48. Owusu Obeng A, Egelund EF, Alsultan A, Peloquin CA, Johnson JA. CYP2C19 polymorphisms and therapeutic drug monitoring of voriconazole: are we ready for clinical implementation of pharmacogenomics? Pharmacotherapy. 2014;34(7):703–18.

    Article  CAS  PubMed  Google Scholar 

  49. Mason NT, Bell GC, Quilitz RE, Greene JN, McLeod HL. Budget impact analysis of CYP2C19-guided voriconazole prophylaxis in AML. J Antimicrob Chemother. 2015;70(11):3124–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Morgan ET. Regulation of cytochrome p450 by inflammatory mediators: why and how? Drug Metab Dispos. 2001;29(3):207–12. Erratum in: Drug Metab Dispos 2001;29(6):932.

    CAS  PubMed  Google Scholar 

  51. Aitken AE, Richardson TA, Morgan ET. Regulation of drug-metabolizing enzymes and transporters in inflammation. Annu Rev Pharmacol Toxicol. 2006;46:123–49.

    Article  CAS  PubMed  Google Scholar 

  52. Veringa A, Ter Avest M, Span LF, van den Heuvel ER, Touw DJ, Zijlstra JG, Kosterink JG, van der Werf TS, Alffenaar JC. Voriconazole metabolism is influenced by severe inflammation: a prospective study. J Antimicrob Chemother. 2017;72(1):261–7.

    Article  CAS  PubMed  Google Scholar 

  53. Chen K, Zhang X, Ke X, Du G, Yang K, Zhai S. Individualized medication of Voriconazole: A Practice Guideline of the Division of Therapeutic Drug Monitoring, Chinese Pharmacological Society. Ther Drug Monit. 2018;40(6):663–74.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Yasu T, Konuma T, Kuroda S, Takahashi S, Tojo A. Effect of cumulative intravenous voriconazole dose on renal function in hematological patients. Antimicrob Agents Chemother. 2018;62(9):e00507–18.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Moriyama B, Obeng AO, Barbarino J, Penzak SR, Henning SA, Scott SA, Agúndez J, Wingard JR, McLeod HL, Klein TE, Cross SJ, Caudle KE, Walsh TJ. Clinical pharmacogenetics implementation Consortium (CPIC) Guidelines for CYP2C19 and voriconazole therapy. Clin Pharmacol Ther. 2017;102(1):45–51.

    Article  CAS  PubMed  Google Scholar 

  56. Yanni SB, Annaert PP, Augustijns P, Ibrahim JG, Benjamin DK Jr, Thakker DR. In vitro hepatic metabolism explains higher clearance of voriconazole in children versus adults: role of CYP2C19 and flavin-containing monooxygenase 3. Drug Metab Dispos. 2010;38(1):25–31.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Pascual A, Csajka C, Buclin T, Bolay S, Bille J, Calandra T, Marchetti O. Challenging recommended oral and intravenous voriconazole doses for improved efficacy and safety: population pharmacokinetics-based analysis of adult patients with invasive fungal infections. Clin Infect Dis. 2012;55(3):381–90.

    Article  CAS  PubMed  Google Scholar 

  58. Wang T, Chen S, Sun J, Cai J, Cheng X, Dong H, Wang X, Xing J, Dong W, Yao H, Dong Y. Identification of factors influencing the pharmacokinetics of voriconazole and the optimization of dosage regimens based on Monte Carlo simulation in patients with invasive fungal infections. J Antimicrob Chemother. 2014;69(2):463–70.

    Article  CAS  PubMed  Google Scholar 

  59. Hamada Y, Tokimatsu I, Mikamo H, Kimura M, Seki M, Takakura S, Ohmagari N, Takahashi Y, Kasahara K, Matsumoto K, Okada K, Igarashi M, Kobayashi M, Mochizuki T, Nishi Y, Tanigawara Y, Kimura T, Takesue Y. Practice guidelines for therapeutic drug monitoring of voriconazole: a consensus review of the Japanese Society of Chemotherapy and the Japanese Society of Therapeutic Drug Monitoring. J Infect Chemother. 2013;19(3):381–92.

    Article  PubMed  PubMed Central  Google Scholar 

  60. Warris A, Lehrnbecher T, Roilides E, Castagnola E, Brüggemann RJM, Groll AH. ESCMID-ECMM guideline: diagnosis and management of invasive aspergillosis in neonates and children. Clin Microbiol Infect. 2019;25(9):1096–113.

    Article  CAS  PubMed  Google Scholar 

  61. Ashbee HR, Barnes RA, Johnson EM, Richardson MD, Gorton R, Hope WW. Therapeutic drug monitoring (TDM) of antifungal agents: guidelines from the British Society for Medical Mycology. J Antimicrob Chemother. 2014;69(5):1162–76.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

This study was supported by the Natural Science Foundation of Hunan Province (2024JJ8227), and the Scientific Research Project of Hunan Provincial Health Commission (W20243243) and Scientific Research Project of Changsha Municipal Health Commission (KJ-B2023042). The funding sources did not have a role in the design, conduct, or analysis of the study.

Author information

Authors and Affiliations

Authors

Contributions

LH and YFL screened the literature and collected the data, LH wrote the manuscript, JJH and GFH revised the manuscript. All authors read and approved the final manuscript.

Corresponding authors

Correspondence to Lin Hu or Gefei He.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing of interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Hu, L., Huang, J., Li, Y. et al. Clinical application of voriconazole in pediatric patients: a systematic review. Ital J Pediatr 50, 113 (2024). https://doi.org/10.1186/s13052-024-01684-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13052-024-01684-z

Keywords