ABSTRACT
Voriconazole (VRC), a first-line agent for the treatment of invasive fungal infections, is mainly metabolized by human cytochrome P450 (CYP) 2C19. In this study, a retrospective analysis was performed to investigate the key factors that influence the plasma trough concentration (Cmin) of VRC, and an appropriate dosing regimen for pediatric patients was drafted subsequently. Overall, factors such as age, CYP2C19 phenotype, and combination medication with proton pump inhibitors accounted for 23.4% of variability in dose-normalized Cmin values of VRC by a multiple linear regression analysis. Dose-normalized Cmin values in the poor metabolizers (PMs) and intermediate metabolizers (IMs) were significantly higher than those in extensive metabolizers (EMs) (P < 0.001). To achieve therapeutic Cmin for CYP2C19 ultrarapid metabolizers (UMs) or EMs, patients aged no more than 12 and more than 12 years required doses of 6.53 ± 2.08 and 3.95 ± 0.85 mg/kg of body weight twice daily (P = 0.007). For CYP2C19 PMs or IMs, patients aged under 12 and over 12 years required doses of 5.75 ± 1.73 and 4.23 ± 0.76 mg/kg twice daily, respectively (P = 0.019). Furthermore, coadministration of rifamycin sodium or omeprazole exhibited significant effects on VRC Cmin. Taken together, it is necessary to pay attention to the impact of CYP2C19 phenotype and drug-drug interactions to achieve optimal therapy.
KEYWORDS: pediatric patients, voriconazole, therapeutic drug monitoring, CYP2C19 phenotype, C min
INTRODUCTION
Immunocompromised children are highly susceptible to invasive fungal infections (1). VRC is a triazole antifungal agent recommended as the first choice for treating or preventing invasive aspergillosis (2). Several studies have found that the Cmin of VRC is associated with its efficacy and safety (3, 4). VRC exhibits nonlinear pharmacokinetics, resulting in highly variable Cmin (5, 6). In children, VRC dosing has been particularly challenging due to decreased oral bioavailability, higher elimination capacity, and larger inter- and intrapatient variability compared with adults (7). There is an agreement that children require higher and personalized doses to attain therapeutic Cmin (8). Multiple studies have recommended initial dose ranges of 7 to 9 mg/kg of body weight twice daily in Caucasian children aged 2 to 12 years (9, 10). In China, it suggested that 5 to 7 mg/kg is adequate in children aged <2 years of Asian origin (11). However, VRC has been commonly prescribed for children ≤12 years with a low maintenance dose of 6 mg/kg in our center. Until now, the optimal dose in pediatric patients has not been fully established.
Apart from age, Cmin of VRC can also be affected by genetic polymorphism of CYP2C19. The allele frequencies of CYP2C19*2 and CYP2C19*3 were 38.6% and 5.2% in Han Chinese populations, respectively, which were 2- to 3-fold higher than those in Europeans (12). The frequencies of PM on CYP2C19 are estimated to be 15.8% in patients of Asian origin compared with 2.2% in Caucasians and 13.7% in Chinese subjects (13). The racial differences imply that Chinese people have a high risk of adverse effects because of elevated Cmin. However, limited data are available describing the correlation between CYP2C19 genetic variants and VRC Cmin in pediatric patients (14, 15).
On the other hand, concomitant medication with drug-metabolizing enzyme inducers like rifampin is associated with decreased Cmin of VRC. While the magnitude of this interaction appears to be less than that observed with rifampin, the coadministration of VRC with rifamycin sodium was more common. The cases reported that Cmin of VRC could be maintained within the therapeutic range when coadministered with rifabutin, so that rifabutin could be a substitute for rifampin (16). Whether the use of rifamycin sodium has a clinically significant effect on VRC still requires more evidence. Proton pump inhibitors (PPIs) are able to influence VRC metabolism by competition inhibition against CYP2C19. The magnitude of the interaction with PPIs was greatly influenced by both the type and the dosage (17). To our knowledge, there have been few reports regarding the interaction of VRC and other PPIs other than omeprazole, such as pantoprazole, lansoprazole, and rabeprazole. The clinical implication of this drug interaction needs to be explored further in pediatric patients.
For these goals, this retrospective analysis aimed to investigate key factors that affected VRC Cmin in Chinese pediatric patients, with particular attention to the CYP2C19 phenotype and drug-drug interactions.
RESULTS
Characteristics of patients.
A total of 108 Chinese pediatric patients (39 females and 69 males) were divided in 3 groups according to age (<6 years, 6 to 12 years, and >12 years) (Fig. 1). The characteristics are summarized in Table 1, and there were no significant differences among the three age groups in any of the covariates. Overall, the median age of total patients was 10 years old (range, 0.67 to 17 years), and 61.11% of patients were younger than 12 years old. The median body weight during VRC treatment was 32.35 kg (range, 7.00 to 84.00 kg). Hematologic malignancies and HSCT were the most common underlying conditions in patients receiving VRC. VRC was administered either targeted therapeutically (n = 27), diagnostic-driven therapeutically (n = 41), empirically (n = 27), or prophylactically (n = 13). CYP2C19 phenotype classifications were UM (n = 1), EM (n = 21), IM (n = 26), and PM (n = 8).
FIG 1.
Flow chart of the study.
TABLE 1.
Characteristics of patients
Parameter | Value for: |
P value | |||
---|---|---|---|---|---|
All patients | <6 yr | 6–12 yr | >12 yr | ||
No. of patients | 108 | 28 | 38 | 42 | |
Age (yr) | 10.50 (0.67–18.00) | 3.00 (0.67–5.00) | 9.00 (6.00–12.00) | 16.00 (13.00–17.00) | |
Body wt (kg) | 32.35 (7.00–84.00) | 14.50 (7.00–24.20) | 29.00 (17.40–46.00) | 55.00 (33.60–84.00) | |
Gender [no. (%)] | 0.630 | ||||
Female | 39 (36.11) | 9 | 16 | 14 | |
Male | 69 (63.89) | 19 | 22 | 28 | |
Underlying disease [no. (%)] | 0.054a | ||||
Acute lymphoblastic | 27 (25.00) | 10 | 8 | 9 | |
Acute myeloid leukemia | 24 (22.22) | 6 | 13 | 5 | |
Acute myelocytic leukemia | 1 (0.93) | 1 | 0 | 0 | |
HSCT | 24 (22.22) | 3 | 11 | 10 | |
Aplastic anemia | 14 (12.96) | 5 | 5 | 4 | |
Others | 18 (16.67) | 3 | 1 | 14 | |
Treatment indication [no. (%)] | 0.484b | ||||
Prophylaxis | 13 (12.04) | 5 | 0 | 8 | |
Empirical | 27 (25.00) | 5 | 17 | 5 | |
Diagnostic-driven therapy | 41 (37.96) | 9 | 15 | 17 | |
Targeted therapy | 27 (25.00) | 9 | 6 | 12 | |
Metabolizer phenotype [no. (%)] | 0.523c | ||||
UM | 1 (0.93) | 0 | 1 | 0 | |
EM | 21 (19.44) | 4 | 9 | 8 | |
IM | 26 (24.07) | 7 | 10 | 9 | |
PM | 8 (7.41) | 3 | 1 | 4 | |
Unknown | 52 (48.15) | 14 | 17 | 21 |
Acute myelocytic leukemia, HSCT, aplastic anemia, and others groups were combined for statistical analysis. HSCT, hematopoietic stem cell transplantation.
Prophylaxis and empirical groups were combined for statistical analysis.
UM and EM groups were combined for statistical analysis, and IM and PM groups were combined for statistical analysis.
Analysis of VRC Cmin.
A total of 348 samples were obtained with a median (range) Cmin of 1.26 μg/ml (<0.05 to 11.24). A median of 2 samples (range, 1 to 16) was drawn per patient, of which 63.79% were therapeutic. Figure 2 shows the distribution of measured VRC Cmin and their respective doses. There was a significant difference in the number of VRC Cmin within the therapeutic range between patients ≤12 years and >12 years (106/187 versus 116/161, χ2 = 8.843, P = 0.003). A higher proportion of patients had subtherapeutic Cmin in patients ≤12 years compared with patients >12 years, which means younger children are at risk for treatment failure (62/187 versus 29/161, χ2 = 10.273, P = 0.001). No predictable relationships between dose and Cmin was observed in pediatric patients (n = 348, coefficient = −0.056, P = 0.295).
FIG 2.
Scatterplot of the VRC Cmin versus the VRC dose. The green square, blue triangles, and orange circles represent data of children aged <6 years, 6 to 12 years, and >12 years, respectively. The area between the horizontal lines indicates the therapeutic range (0.5 to 5.0 μg/ml).
Factors influencing dose-normalized VRC Cmin.
The concentrations of VRC in pediatric patients were affected by multiple factors, including age, CYP2C19 genetic polymorphism, drug interactions, and so on (Table 2). Multivariate analysis by linear regression was performed to identify the factors associated with VRC Cmin corrected for dose (Table 3). No significant effect was detected of routes of administration, glutamyl transpeptidase, or AKP on VRC Cmin corrected for dose, but age, CYP2C19 phenotype, and PPI comedication did have a significant impact.
TABLE 2.
Univariate analysis of factors influencing dose normalized plasma VRC concentration
Covariate | Coeffcient | P valuea |
---|---|---|
Age | 0.380 | <0.001 |
Body wt | 0.393 | <0.001 |
Gender | ||
Female | 0.175 | |
Male | ||
CYP2C19 phenotype | ||
UM/EM | <0.001 | |
IM/PM | ||
PPIs | ||
Not concomitantly administered | 0.016 | |
Concomitantly administered | ||
Routes of administration | ||
Injection | 0.133 | |
Oral | ||
ALT | −0.111 | 0.143 |
AST | −0.071 | 0.350 |
Glutamyl transpeptidase | 0.180 | 0.017 |
AKP | −0.163 | 0.031 |
Bilirubin | 0.116 | 0.138 |
The boldface values indicate significant differences (P < 0.05).
TABLE 3.
Multiple linear regression analysis for dose-corrected VRC Cmin
Covariate | Coeffcient | 95% CI | P value | R 2 |
---|---|---|---|---|
Age | 0.437 | 0.029, 0.056 | <0.001 | 0.234 |
CYP2C19 phenotype | ||||
UM/EM | 0.192 | 0.058, 0.337 | 0.006 | |
IM/PM | ||||
PPIs | ||||
Not concomitantly administered | −0.182 | −0.309, −0.043 | 0.01 | |
Concomitantly administered | ||||
Routes of administration | ||||
Injection | 0.107 | −0.042, 0.296 | 0.14 | |
Oral |
Impact of CYP2C19 phenotype on dose-normalized Cmin.
A scatterplot of the relationships between the CYP2C19 phenotype and Cmin corrected for dose is shown in Fig. 3. CYP2C19 IMs (median, 0.31 μg/ml/mg/kg; range, 0.01 to 2.60; P < 0.001) and PMs (median, 0.48 μg/ml/mg/kg; range, 0.04 to 1.75; P < 0.001) had higher dose-normalized Cmin than EMs (median, 0.11 μg/ml/mg/kg; range, 0.01 to 2.02). CYP2C19 UMs did not have significantly lower dose-normalized Cmin (median, 0.09 μg/ml/mg/kg; range, 0.04 to 0.60; P = 1.000) than EMs. There was no difference in the dose-normalized Cmin between the CYP2C19 PMs and IMs (P = 0.155).
FIG 3.
CYP2C19 phenotype and dose-normalized Cmin. Horizontal bars represent median values and interquartile range of dose-normalized Cmin.
Maintenance dose to achieve the target range.
Patients whose Cmin were within therapeutic range were divided into four groups according to age and phenotype, and the average maintenance dose required to achieve therapeutic Cmin was inversely related to age (Fig. 4). Patients with CYP2C19 UM or EM phenotype of ≤12 and >12 years required mean ± standard deviation (SD) doses of 6.53 ± 2.08 and 3.95 ± 0.85 mg/kg twice daily, respectively (P = 0.007). With CYP2C19 PM or IM phenotype, patients ≤12 and >12 years required doses of 5.75 ± 1.73 and 4.23 ± 0.76 mg/kg twice daily, respectively (P = 0.019), to achieve therapeutic Cmin. In children ≤12 and >12 years, there was no difference in the EM/UM and PM/IM groups required to achieve therapeutic Cmin (≤12 years, 6.53 ± 2.08 mg/kg versus 5.75 ± 1.73 mg/kg, P = 0.274; >12 years, 3.95 ± 0.85 mg/kg versus 4.23 ± 0.76 mg/kg, P = 0.505). Before genotyping of CYP2C19, the previous pediatric dosing regimen was explored according to therapeutic drug monitoring and clinical experience, leading to attainment of target concentrations of 56.68% in children ≤12 years. No significant differences between the doses recommended for each CYP2C19 phenotype/age group and the previous dose were observed.
FIG 4.
Average dose required to achieve target VRC Cmin (0.5 to 5 μg/ml) categorized by age and CYP2C19 phenotype. Horizontal bars represent mean ± SD values of VRC Cmin.
Influence of drug interactions on VRC Cmin.
Medicines for possible drug interaction with VRC were identified. More than 59.26% of patients (64/108) had concomitant medications with potential drug interaction with VRC: rifamycin sodium (n = 17), rifampin (n = 1), fluconazole (n = 1), omeprazole (n = 32), pantoprazole (n = 39), lansoprazole (n = 16), and rabeprazole (n = 4). According to clinical experience, when patients received concomitant rifamycin sodium, VRC Cmin were obviously low until 14 days after discontinuation. As shown in Fig. 5a, there was a statistically significant difference in dose-normalized VRC Cmin between the groups coadministered with or without rifamycin sodium (P < 0.001). Moreover, the patients receiving PPIs had a higher dose-normalized VRC Cmin (P = 0.038). Of the five PPIs, omeprazole was shown to have a significant effect on VRC concentration (Fig. 5b) (P < 0.001) compared with those with no PPIs, whereas pantoprazole (P = 0.870), lansoprazole (P = 0.828), and rabeprazole (P = 0.208) were not found to influence VRC Cmin significantly. VRC Cmin was significantly lower in patients treated with pantoprazole (P = 0.006) and lansoprazole (P = 0.023) versus those treated with omeprazole. There were no significant differences in VRC Cmin of patients with omeprazole and rabeprazole (P = 0.711).
FIG 5.
(a) Effect of rifamycin sodium on dose-normalized Cmin. (b) Effect of omeprazole on dose-normalized Cmin. Horizontal bars represent median values of dose-normalized Cmin.
Relationship between VRC Cmin and treatment response.
Outcome data were analyzed in patients with targeted antifungal therapy. Data were available for 27 courses. Twenty-one had a successful outcome (21/27; 77.78%), while six were classified as failure (6/27; 22.22%). Univariate analysis did not reveal any statistically significant association between mean Cmin values and treatment response (P = 0.369). The incidence of treatment success in all patients was 88.89% (96/108). No association between Cmin values and treatment response was found (P = 0.158), although 20.37% (22/108) of patients did not attain mean Cmin of ≥0.5 μg/ml.
According to the CTCAE criteria, 18 patients (16.67%) exhibited hepatotoxicity 14 days (range, 2 to 26 days) after starting VRC therapy. Hepatotoxicity was more common in patients with higher Cmin (1.73 μg/ml versus 1.04 μg/ml, P = 0.013) (Fig. 6). Logistic regression analysis showed that the probability of hepatotoxicity increased by 16.2% while Cmin increased by 1 μg/ml. When Cmin is higher than 2.4 μg/ml, the incidence of hepatotoxicity is significantly increased (χ2 = 3.938, P = 0.047). Other adverse drug reactions were observed in four pediatric patients (3.7%) during VRC treatment, including skin rashes in one case, myocardial injury in two cases, and neutropenia in one case. No ophthalmological disorders were reported, likely due to the young age of the patients.
FIG 6.
Hepatotoxicity and VRC Cmin. Horizontal bars represent median, minimum, and maximum values of VRC Cmin.
DISCUSSION
For children, maintaining VRC Cmin within a target range is clinically important in order to obtain an optimal therapeutic response. We found high interindividual variability in VRC Cmin that cannot be explained by a dose-concentration relationship. A large fraction of pediatric patients fails to achieve therapeutic Cmin, with a consequently increased risk of therapeutic failure. This study demonstrated that serum concentrations of VRC were affected by age, CYP2C19 phenotype, and coadministration of PPIs (Table 3).
As has been shown previously in adults (18, 19), results of the current study indicate that VRC Cmin in pediatric patients is correlated with CYP2C19 polymorphisms. CYP2C19 IMs and PMs had higher dose-normalized Cmin than EMs, and there was no difference in the dose-normalized Cmin between the CYP2C19 PMs and IMs (Fig. 3). In addition, the incidence of IM/PM among Asian populations, about 60.71% in our data, is higher than that for Caucasians. Therefore, it remains to be considered whether the recommended dose (7 to 9 mg/kg intravenously every 12 h) of previous studies conducted in western nations should be used in China. Our findings suggest that, in the younger age group, the recommended dose should be increased. For CYP2C19 UMs or EMs, most children ≤12 and >12 years should receive maintaining doses of 6.53 and 3.95 mg/kg twice daily, respectively; with CYP2C19 PM or IM phenotype, patients ≤12 and >12 years, respectively, required doses of 5.75 and 4.23 mg/kg twice daily. Lower VRC doses were required to achieve the target Cmin among older children, which was consistent with one previous study (20).
Doses based on age and CYP2C19 phenotype should theoretically improve attainment of target concentrations, but there were no significant differences in the required dose and the previous dose. Our data cannot provide the guidance for optimizing VRC dosing based on the phenotype. The most likely cause is the small sample size for each phenotype group in this retrospective study, but it was suggested that phenotype should be identified before dosing. There was a significant difference in Cmin between different phenotype groups. CYP2C19 phenotype to predict Cmin may be a strategy for reducing the risk of subtherapeutic and supratherapeutic Cmin.
Rifamycin sodium may upregulate CYP2C19 expression, thereby resulting in decreased VRC Cmin, even lower than the lower limit of quantification of 0.05 μg/ml. The findings suggest that rifamycin sodium’s inductive effects were notable for at least 14 days. Patients could benefit from the selection of other drugs instead of rifamycin sodium to avoid the risk of subtherapeutic VRC Cmin and therapeutic failure. This retrospective study is the first to assess the influence of comedications with rifamycin sodium in children.
Notably, out of the five most frequently recorded concomitant PPIs, omeprazole promoted higher dose-normalized VRC Cmin. In contrast, pantoprazole, lansoprazole, and rabeprazole showed no significant effects on VRC Cmin. A multicenter prospective study in adults has demonstrated increased VRC exposure with omeprazole due to an inhibition of CYP2C19 (21). Our data support differences in the degrees of interaction observed with different PPIs. This observation is consistent with the results of a physiologically based pharmacokinetic model that pharmacokinetic values of VRC increased to various degrees when administered PPIs (22). There was a statistically significant association between increased VRC Cmin and omeprazole use but not with rabeprazole, lansoprazole, or pantoprazole use, suggesting a greater CYP2C19 enzyme inhibitory effect of omeprazole. That should be helpful for the choice of PPIs for pediatric patients treated with VRC. There were no significant differences in VRC Cmin of patients with omeprazole and rabeprazole, which may be due to the low number of patients with rabeprazole.
We found no consistent correlation between VRC mean Cmin values and treatment response, similar to findings of Liu et al. (11). A large portion of patients used VRC as diagnostic-driven therapy, empirical therapy, or prophylaxis. They showed clinical improvement regardless of Cmin, which might explain why VRC did not show significant improvement in treatment success rates. Second, antifungal combination therapy with echinocandins or amphotericin B may confound the assessment of an exposure-response relationship of VRC.
Hepatotoxicity was the most frequent adverse event, and the incidence (16.67%) is comparable to previously reported rates (14.3% to 17.5%) (23, 24). A Cmin of >2.4 μg/ml is associated with increased hepatotoxicity, which is considerably lower than that described in previous studies (25). The relationship between VRC Cmin and hepatotoxicity may follow a different profile among pediatric patients. A meta-analysis indicated that the optimal trough concentration for increasing clinical success and minimizing hepatotoxicity during VRC therapy for Asian children populations is 1.0 to 3.0 μg/ml (26). Therefore, a lowered upper limit of the target Cmin should be considered for Asian pediatric patients compared with adults.
However, some limitations are present in this study. This was a retrospective analysis of VRC drug monitoring from a single tertiary center in China. We failed to find a positive relationship between VRC Cmin and treatment response. Despite these limitations, this study supports several conclusions regarding clinical application of VRC in pediatric patients: (i) the CYP2C19 genotype is likely one of the factors contributing to the high variability of VRC Cmin in pediatric patients, and (ii) coadministration of rifamycin sodium or omeprazole may interact with VRC, resulting in significantly lower or higher plasma Cmin.
As a result, we suggest that CYP2C19 phenotype helps in explaining lower VRC dosage required in Chinese populations, considering a higher portion of poor CYP2C19 metabolizers than those for Caucasian or African-American populations (27). A careful selection of the drugs that are concomitantly administered with VRC should be conducted to obtain an optimal clinical outcome. A prospective study with larger patient numbers is warranted to confirm the results of the present study, which showed the association between VRC Cmin and CYP2C19 phenotype in pediatric patients.
MATERIALS AND METHODS
Patients and data collection.
A retrospective observational study of pediatric patients below the age of 18 who were subjected to VRC Cmin monitoring between May 2017 and October 2020 was performed (Fig. 1). Clinical and demographic data were collected from the electronic medical records in The First Affiliated Hospital of Zhengzhou University (Zhengzhou, China). This study was approved by the Zhengzhou University Medical Research and Ethics Committee (number 2020-KY-0355-002). Demographic characteristics, drug administration history, outcomes of therapy, adverse events, and laboratory test results, including alanine aminotransferase (ALT), aspartate aminotransferase (AST), glutamyl transpeptidase, alkaline phosphatase (AKP), and bilirubin, were also collected. In addition, rifampin and rifamycin sodium can significantly influence VRC Cmin even for 2 weeks after discontinuation (28, 29). Therefore, the biological data were collected on the day Cmin was measured, and potential drug interactions with rifampin or rifamycin sodium were collected during the 14 days before Cmin measurements. Only patients with a VRC treatment regimen of ≥14 days were used to assess clinical response.
Diagnostic criteria.
Diagnosis and treatment of invasive fungal infections was classified according to the definitions of the European Organization for Research and Treatment of Cancer/Mycoses Study Group (EORTC/MSG). Treatment response was assessed by clinical, radiological, and microbiological criteria. Outcome for targeted therapy was classified as a success when two of the following criteria were met: clinical improvement, radiologic improvement, or decreasing mycologic burden. Otherwise, the course was defined as a failure. Diagnostic-driven, empirical therapy and prophylaxis were classified as a success if the course was completed without breakthrough fungal infection (30). Adverse drug reactions and their relationship with VRC were based on the Common Terminology Criteria for Adverse Events (CTCAE) defined by the National Cancer Institute.
Therapeutic drug monitoring of VRC.
VRC Cmin between 0.5 and 5 μg/ml were considered therapeutic. Only patients having achieved steady state were included in the analysis, and for patients who received a loading dose, Cmin measurements taken on day 3 of dosing or later were included. In patients who did not receive a loading dose, Cmin measurements taken on day 5 of dosing or later were included. Dose adjustments were taken according to the guidelines (31).
Cmin was measured by an ultra-high-performance liquid chromatograph-tandem mass spectrometry method (UPLC-MS/MS; Waters, United States). Following protein precipitation, the analyte and internal standard voriconzole-d3 (TRC; purity was 98%) were separated on a C18 column (Waters Acquity UPLC BEH; 2.1 mm by 50 mm, 1.7 μm). The electrospray ion source (ESI) was employed as the ion source. MS acquisition was performed in multiple reaction monitoring mode, and the mass transitions were 350.0→127.0 for VRC and 353.0→127.0 for internal standard. This method was developed and validated according to FDA guidelines. The analytical range was 0.05 to 10.00 μg/ml. The intra- and interassay relative standard deviations (RSD) were 3.43 to 6.18%, 4.47 to 4.55%, and 2.54 to 3.28% for low, medium, and high levels of internal quality controls, respectively. The intra- and interassay relative deviation (RE) was ≤4.66%. The accepted inter/intraday precision and accuracy were less than 20%.
CYP2C19 phenotype assignment.
Genotyping was performed at the Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou University, using the sequencing-by-hybridization array on a Fascan 48E (Xi'an Tianlong Technology Co.). Patients were classified into categories of metabolizer phenotypes with the use of the established common-consensus star allele nomenclature. Thus, carriers of a single CYP2C19 *17 allele (*1/*17) and *17 homozygotes were classified as UMs; patients without a CYP2C19 *2, *3, or *17 allele (*1/*1) were classified as EMs; those with one CYP2C19*2 or *3 allele (*1/*2 or *1/*3) and patients with one CYP2C19 *17 allele and one loss-of-function allele (*2/*17 or *3/*17) were classified as IMs; and those with two CYP2C19 *2 or *3 alleles (*2/*2, *2/*3, or *3/*3) were classified as PMs (32).
Data analysis.
IBM SPSS Statistics software v.19.0 (IBM Corp., Armonk, NY) was used for statistical analysis. The χ2 test was used to compare patient characteristics between age groups. Univariate analysis of factors influencing dose-normalized plasma VRC concentration was conducted using the Spearman correlation analysis or Mann-Whitney U. Variables with P values of <0.05 by univariate analysis were entered into the multivariable model. The correlation between covariates (age, CYP2C19 phenotype, interacting drugs, and routes of administration) and VRC Cmin corrected for dose were examined using multiple linear regression analysis. The association of Cmin with efficacy and safety was analyzed by logistic regression. Continuous variables were compared by t test or nonparametric Mann-Whitney U test. A P value of <0.05 was considered statistically significant.
REFERENCES
- 1.Pana ZD, Roilides E, Warris A, Groll AH, Zaoutis T. 2017. Epidemiology of invasive fungal disease in children. J Pediatric Infect Dis Soc 6:S3–S11. 10.1093/jpids/pix046. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Lestrade PP, Bentvelsen RG, Schauwvlieghe AFAD, Schalekamp S, Velden W, Kuiper EJ, Paassen J, Hoven B, Lee HA, Melchers WJG, Haan AF, Hoeven HL, Rijnders BJA, Beek MT, Verweij PE. 2019. Voriconazole resistance and mortality in invasive aspergillosis: a multicenter retrospective cohort study. Clin Infect Dis 68:1463–1471. 10.1093/cid/ciy859. [DOI] [PubMed] [Google Scholar]
- 3.Jin HY, Wang TS, Falcione BA, Olsen KM, Chen K, Tang HL, Hui J, Zhai S. 2016. Trough concentration of voriconazole and its relationship with efficacy and safety: a systematic review and meta-analysis. J Antimicrob Chemother 71:1772–1785. 10.1093/jac/dkw045. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Bellmann R, Smuszkiewicz P. 2017. Pharmacokinetics of antifungal drugs: practical implications for optimized treatment of patients. Infection 45:737–779. 10.1007/s15010-017-1042-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Stott KE, Hope WW. 2017. Therapeutic drug monitoring for invasive mould infections and disease: pharmacokinetic and pharmacodynamic considerations. J Antimicrob Chemother 72:i12–i18. 10.1093/jac/dkx029. [DOI] [PubMed] [Google Scholar]
- 6.Carlesse FADMC, de Araujo OR, Marques LMA, da Silva DCB, Senerchia AA, Petrilli AS. 2019. A pharmacokinetic model for voriconazole in a highly diversified population of children and adolescents with cancer. Mycoses 62:399–404. 10.1111/myc.12899. [DOI] [PubMed] [Google Scholar]
- 7.Kang S, Yee J, Kim JY, Han HW, Kang SO, Lee KE, Gwak HS. 2020. Factors associated with voriconazole concentration in pediatric patients. Ther Drug Monit 42:866–871. 10.1097/FTD.0000000000000804. [DOI] [PubMed] [Google Scholar]
- 8.Kang HM, Lee HJ, Cho EY, Yu K-S, Lee H, Lee JW, Kang HJ, Park KD, Shin HY, Choi EH. 2015. The clinical significance of voriconazole therapeutic drug monitoring in children with invasive fungal infections. Pediatr Hematol Oncol 32:557–567. 10.3109/08880018.2015.1088905. [DOI] [PubMed] [Google Scholar]
- 9.Bartelink IH, Wolfs T, Jonker M, De Waal M, Egberts TCG, Ververs TT, Boelens JJ, Bierings M. 2013. Highly variable plasma concentrations of voriconazole in pediatric hematopoietic stem cell transplantation patients. Antimicrob Agents Chemother 57:235–240. 10.1128/AAC.01540-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Zembles TN, Thompson NE, Havens PL, Kaufman BA, Huppler AR. 2016. An optimized voriconazole dosing strategy to achieve therapeutic serum concentrations in children younger than 2 years old. Pharmacotherapy 36:1102–1108. 10.1002/phar.1829. [DOI] [PubMed] [Google Scholar]
- 11.Liu L, Zhou X, Wu TT, Jiang HL, Yang ST, Zhang Y. 2017. Dose optimisation of voriconazole with therapeutic drug monitoring in children: a single-centre experience in China. Int J Antimicrob Agents 49:483–487. 10.1016/j.ijantimicag.2016.11.028. [DOI] [PubMed] [Google Scholar]
- 12.Zuo LJ, Guo T, Xia DY, Jia LH. 2012. Allele and genotype frequencies of CYP3A4, CYP2C19, and CYP2D6 in Han, Uighur, Hui, and Mongolian Chinese populations. Genet Test Mol Biomarkers 16:102–108. 10.1089/gtmb.2011.0084. [DOI] [PubMed] [Google Scholar]
- 13.Shimizu T, Ochiai H, Asell F, Shimizu H, Saitoh R, Hama Y, Katada J, Hashimoto M, Matsui H, Taki K, Kaminuma T, Yamamoto M, Aida Y, Ohashi A, Ozawa N. 2003. Bioinformatics research on inter-racial difference in drug metabolism I. Analysis on frequencies of mutant alleles and poor metabolizers on CYP2D6 and CYP2C19. Drug Metab Pharmacokinet 18:48–70. 10.2133/dmpk.18.48. [DOI] [PubMed] [Google Scholar]
- 14.Blanco-Dorado S, Maroñas O, Latorre-Pellicer A, Jato MTR, López-Vizcaíno A, Márquez AG, García BB, Medall DB, Castiñeiras GB, Bernal M, Campos-Toimil M, Espinar FO, Hortas AB, Piñeiro GD, Ferro IZ, Carracedo Á, Lamas MJ, Fernández-Ferreiro A. 2020. Impact of CYP2C19 genotype and drug interactions on voriconazole plasma concentrations: a spain pharmacogenetic‐pharmacokinetic prospective multicenter study. Pharmacotherapy 40:17–25. 10.1002/phar.2351. [DOI] [PubMed] [Google Scholar]
- 15.Hamadeh IS, Klinker KP, Borgert SJ, Richards AI, Li W, Mangal N, Hiemenz JW, Schmidt S, Langaee TY, Peloquin CA, Johnson JA, Cavallari LH. 2017. Impact of the CYP2C19 genotype on voriconazole exposure in adults with invasive fungal infections. Pharmacogenet Genomics 27:190–196. 10.1097/FPC.0000000000000277. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Schwiesow JN, Iseman MD, Peloquin CA. 2008. Concomitant use of voriconazole and rifabutin in a patient with multiple infections. Pharmacotherapy 28:1076–1080. 10.1592/phco.28.8.1076. [DOI] [PubMed] [Google Scholar]
- 17.Cojutti P, Candoni A, Forghieri F, Isola M, Zannier ME, Bigliardi S, Luppi M, Fanin R, Pea F. 2016. Variability of voriconazole trough levels in haematological patients: influence of comedications with cytochrome P450(CYP) inhibitors and/or with CYP inhibitors plus CYP Inducers. Basic Clin Pharmacol Toxicol 118:474–479. 10.1111/bcpt.12530. [DOI] [PubMed] [Google Scholar]
- 18.Hicks JK, Quilitz RE, Komrokji RS, Kubal TE, Lancet JE, Pasikhova Y, Qin D, So W, Caceres G, Kelly K, Salchert YS, Shahbazian K, Abbas-Aghababazadeh F, Fridley BL, Velez AP, McLeod HL, Greene JN. 2020. Prospective CYP2C19‐guided voriconazole prophylaxis in patients with neutropenic acute myeloid leukemia reduces the incidence of subtherapeutic antifungal plasma concentrations. Clin Pharmacol Ther 107:563–570. 10.1002/cpt.1641. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Hope W, Johnstone G, Cicconi S, Felton T, Goodwin J, Whalley S, Santoyo-Castelazo A, Ramos-Martin V, Lestner J, Credidio L, Dane A, Carr DF, Pirmohamed M, Salim R, Neely M. 2019. Software for dosage individualization of voriconazole: a prospective clinical study. Antimicrob Agents Chemother 63:e02353-18. 10.1128/AAC.02353-18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Takahashi T, Smith AR, Jacobson PA, Fisher J, Rubin NT, Kirstein MN. 2020. Impact of obesity on voriconazole pharmacokinetics among pediatric hematopoietic cell transplant recipients. Antimicrob Agents Chemother 64:e00653-20. 10.1128/AAC.00653-20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Dorado SB, Amigo OM, Latorre-Pellicer A, Jato MTR, López-Vizcaíno A, Márquez AG, García BB, Medall DB, Castiñeiras GB, Bernal M, Campos-Toimil M, Espinar FO, Hortas AB, Ferro IZ, Carracedo Á, Lamas MJ, Fernández-Ferreiro A. 2020. A multicentre prospective study evaluating the impact of proton-pump inhibitors omeprazole and pantoprazole on voriconazole plasma concentrations. Br J Clin Pharmacol 86:1661–1666. 10.1111/bcp.14267. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Qi F, Zhu L, Li N, Ge T, Xu G, Liao S. 2017. Influence of different proton pump inhibitors on the pharmacokinetics of voriconazole. Int J Antimicrob Agents 49:403–409. 10.1016/j.ijantimicag.2016.11.025. [DOI] [PubMed] [Google Scholar]
- 23.Boast A, Curtis N, Cranswick N, Gwee A. 2016. Voriconazole dosing and therapeutic drug monitoring in children: experience from a paediatric tertiary care centre. J Antimicrob Chemother 71:2031–2036. 10.1093/jac/dkw056. [DOI] [PubMed] [Google Scholar]
- 24.Mori M, Kobayashi R, Kato K, Maeda N, Fukushima K, Goto H, Inoue M, Muto C, Okayama A, Watanabe K, Liu P. 2015. Pharmacokinetics and safety of voriconazole intravenous-to-oral switch regimens in immunocompromised Japanese pediatric patients. Antimicrob Agents Chemother 59:1004–1013. 10.1128/AAC.04093-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Hamada Y, Ueda T, Miyazaki Y, Nakajima K, Fukunaga K, Miyazaki T, Nakada-Motokawa N, Nagao M, Kawamura H, Shigemi A, Ebihara F, Kimura T, Ikegame K, Uchino M, Ikeuchi H, Takesue Y. 2020. Effects of antifungal stewardship using therapeutic drug monitoring in voriconazole therapy on the prevention and control of hepatotoxicity and visual symptoms: a multicentre study conducted in Japan. Mycoses 63:779–786. 10.1111/myc.13129. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Hanai Y, Hamada Y, Kimura T, Matsumoto K, Takahashi Y, Fujii S, Nishizawa K, Takesue Y. 2021. Optimal trough concentration of voriconazole with therapeutic drug monitoring in children: a systematic review and meta-analysis. J Infect Chemother 27:151–160. 10.1016/j.jiac.2020.11.014. [DOI] [PubMed] [Google Scholar]
- 27.Dean L. 2012. Clopidogrel therapy and CYP2C19 genotype. InPratt VM, Scott SA, Pirmohamed M, Esquivel B, Kane MS, Kattman BL, Malheiro AJ (ed), Medical genetics summaries. National Center for Biotechnology Information, Bethesda, MD. https://www.ncbi.nlm.nih.gov/books/NBK84114/. [PubMed] [Google Scholar]
- 28.Baciewicz AM, Chrisman CR, Finch CK, Self TH. 2013. Update on rifampin, rifabutin, and rifapentine drug interactions. Curr Med Res Opin 29:1–12. 10.1185/03007995.2012.747952. [DOI] [PubMed] [Google Scholar]
- 29.Farrokh S, Avdic E. 2019. Voriconazole autoinduction and saturable metabolism after cessation of rifampin in a patient with invasive central nervous system Aspergillus: importance of therapeutic drug monitoring. J Pharm Pract 32:589–594. 10.1177/0897190018760621. [DOI] [PubMed] [Google Scholar]
- 30.Anonymous. 2020. The Chinese guidelines for the diagnosis and treatment of invasive fungal disease in patients with hematological disorders and cancers (the 6th revision). Zhonghua Nei Ke Za Zhi 59:754–763. [DOI] [PubMed] [Google Scholar]
- 31.Chen K, Zhang X, Ke X, Du G, Yang K, Zhai S. 2018. Individualized medication of voriconazole: a practice guideline of the division of therapeutic drug monitoring, Chinese pharmacological society. Ther Drug Monit 40:663–674. 10.1097/FTD.0000000000000561. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Waring RH. 2020. Cytochrome P450: genotype to phenotype. Xenobiotica 50:9–18. 10.1080/00498254.2019.1648911. [DOI] [PubMed] [Google Scholar]