Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2016 Feb 23.
Published in final edited form as: Curr Fungal Infect Rep. 2015 Apr 16;9(2):74–87. doi: 10.1007/s12281-015-0219-0

Therapeutic Drug Monitoring and Genotypic Screening in the Clinical Use of Voriconazole

Brad Moriyama 1, Sameer Kadri 2, Stacey A Henning 1, Robert L Danner 2, Thomas J Walsh 3, Scott R Penzak 4
PMCID: PMC4764088  NIHMSID: NIHMS704041  PMID: 26918067

Abstract

Voriconazole is an antifungal triazole that is the first line agent for treatment of invasive aspergillosis. It is metabolized by CYP2C19, CYP2C9, and CYP3A4 and demonstrates wide interpatient variability in serum concentrations. Polymorphisms in CYP2C19 contribute to variability in voriconazole pharmacokinetics. Here, evidence is examined for the use of voriconazole therapeutic drug monitoring (TDM) and the role of CYP2C19 genotyping in voriconazole dosing. The majority of studies exploring the impact of voriconazole TDM on efficacy and safety have found TDM to be beneficial. However, most of these studies are observational, with only one being a randomized controlled trial. High-volume multicenter randomized controlled trials of TDM are currently not available to support definitive guidelines. There is a significant relationship in healthy volunteers between CYP2C19 genotype and voriconazole pharmacokinetics, but this association is markedly less visible in actual patients. While CYP2C19 genotype data may explain variability of voriconazole serum levels, they alone are not sufficient to guide initial dosing. The timeliness of availability of CYP2C19 genotype data in treatment of individual patients also remains challenging. Additional studies are needed before implementation of CYP2C19 genotyping for voriconazole dosing into routine clinical care.

Keywords: voriconazole, CYP2C19, pharmacokinetics, pharmacogenomics

Introduction

Voriconazole is an antifungal triazole approved by the FDA for the treatment of invasive aspergillosis, esophageal candidiasis, candidemia in non-neutropenic patients, disseminated Candida infections, and infections caused by Scedosporium apiospermum and Fusarium spp. [1]. Infectious Diseases Society of America (IDSA) guidelines recommend voriconazole as a first line agent for the treatment of invasive aspergillosis and as an alternative agent for the treatment of candidemia [2,3].

Voriconazole is metabolized in the liver by CYP3A4, CYP2C9 and CYP2C19 and demonstrates wide interpatient variability in serum concentrations [4,5]. Polymorphisms in CYP2C19, but not CYP3A5 or CYP2C9 have been reported to affect its pharmacokinetics [68]. Other factors including age, liver function, and concomitant medications contribute to variability in voriconazole concentrations [4,5]. In addition, voriconazole demonstrates saturable, nonlinear pharmacokinetics in adults [9].

The field of pharmacogenomics seeks to understand variations in the response to medications based on inherited and acquired genetic differences between patients [10,11]. The prospect of pharmacogenomics testing has been explored for a myriad of medications including CYP2C19 genotyping to aid voriconazole dosing [12]. Guidelines from the Royal Dutch Pharmacists Association Pharmacogenetics Working Group, recommend monitoring voriconazole serum concentrations in patients expressing the CYP2C19 poor metabolizer (PM) and CYP2C19 intermediate metabolizer phenotypes [13]. However, there is a paucity of guidelines for pharmacogenomic testing in patients treated with voriconazole. Common side effects of voriconazole include hepatotoxicity, neurotoxicity, blurry vision, skin rash and hyperfluorosis [14]. Some of these adverse effects are more likely to occur at higher than necessary voriconazole serum/plasma concentrations, while low voriconazole levels may result in therapeutic failure [12]. As such, voriconazole therapeutic drug monitoring (TDM) has become commonplace in the management of serious fungal infections. The objective of this review is to critically examine the evidence of the relationships among CYP2C19 genotype, voriconazole serum concentrations, and clinical outcomes.

CYP2C19 polymorphisms

CYP2C19 catalyzes the metabolism of numerous commonly prescribed drugs including antidepressants, anticancer agents, clopidogrel, proton pump inhibitors, diazepam, and voriconazole [1517]. The CYP2C19 gene exhibits significant ethnic differences in expression among the 34 identified alleles [18]. The fully functional allele (*1) is associated with normal CYP2C19 activity; individuals homozygous for this allele are considered extensive metabolizers (EM). Individuals who carry two null alleles are considered poor metabolizers (PMs). The two most common nonfunctional (null) alleles are CYP2C19*2 and CYP2C19*3, which account for 95% of individuals considered PMs [19]. Additional null alleles include CYP2C19*4, CYP2C19*5, CYP2C19*6, and CYP2C19*8 [15,20]. Approximately 3–5% of Caucasians, 12–23% of Asians, 7% of African Americans, and 0.9% of Hispanics are CYP2C19 PMs, indicating that they encode non-functioning enzymes [2124]. Conversely, intermediate metabolizers carry one null and one wild-type allele (i.e. CYP2C19*1/*3).

An allelic variant associated with increased CYP2C19 expression (CYP2C19*17) and catalytic activity has also been identified [25]. Individuals possessing this allelic variant are considered ultra-rapid metabolizers (URM) [17]. CYP2C19*17 is a relatively common allele in Europeans and Africans (18–27% and 10–26% frequencies, respectively), yet it occurs infrequently in Asians (0.15 – 0.44% prevalence) [17].

Voriconazole concentrations and efficacy/toxicity

Voriconazole exhibits a narrow therapeutic index, non-linear pharmacokinetics, marked genotypic variability in CYP2C19 metabolizer status and a high propensity for drug-drug interactions [5,26]. Consequently, a fixed dose of voriconazole yields a myriad of plasma concentrations [2729], which do not necessarily predict future concentrations even in the same individual [30,31]. Voriconazole is also commonly used for fungal infections such as invasive aspergillosis that are associated with significant mortality among susceptible hosts. These factors underscore the need for voriconazole TDM. Indeed, the majority of studies that explore the impact of voriconazole TDM on efficacy and safety have found it to be beneficial. However, most of these studies are observational, with only one being a randomized, assessor-blinded, controlled trial [32]. Furthermore, voriconazole TDM studies exhibit marked heterogeneity in quality and design, including assay techniques, voriconazole sampling methodology [29,32,33], and target concentration ranges, all of which pose difficulties when comparing results across studies. Nonetheless, the IDSA guidelines support the use of voriconazole TDM [3].

Studies examining the role of voriconazole TDM have used a variety of voriconazole-exposure metrics. Most of the early studies reported random blood sampling. Voriconazole efficacy has been shown to correlate well in vivo with the AUC/MIC [34,35] or mean unbound voriconazole concentration/MIC ratio [36]. However, these measurements entail repeated sampling and are difficult to perform in the clinical setting. Assessment of the trough/MIC ratio is a less robust, but more clinically achievable approach, which was found to predict clinical response in a 5000-patient Monte Carlo simulation using data from multiple Phase 2 and Phase 3 clinical trials [36].

The optimal target concentration range for voriconazole is not clearly defined. The suggested lower end cut-off for efficacy has ranged widely between 0.25 mcg/mL [33] and 2.51 mcg/mL [37]. Most initial studies used a voriconazole concentration > 1 mcg/mL as the lower cut-off on the basis of in vitro studies that reported voriconazole MICs between 0.5 and 1 mcg/mL for most Aspergillus spp. and Candida spp. [38,39]. However, data now suggest a voriconazole concentration of 2 mcg/mL may be a more appropriate lower-end threshold concentration [4042]. Because the unbound circulating fraction (40–50%) of voriconazole is microbiologically active [29], such a recommendation seems pharmacologically plausible. Suggested upper-end threshold concentrations for voriconazole range from 4 to 7 mcg/mL and are based on concentrations above which toxicities were observed across studies. Studies reporting relationships among voriconazole concentrations and efficacy and toxicity are summarized in Table 1 and discussed in detail below [29,3133,36,4350].

Table 1.

Studies Reporting the Relationship Between Voriconazole Concentrations and Efficacy and/or Toxicityi

Study Design Sample Size Population TDM Efficacy Relationship TDM Toxicity Relationship Suggested Thresholds (mcg/mL) Ref.
Lower Upper
RCT (Assessor blinded; Single center) 110 (1:1 randomization) Patients with IFIs receiving targeted or empiric therapy Higher success in TDM groupa (86% vs. 63%; p=0.04) No difference in AE incidence, but lesser drug discontinuations in the TDM group (4% vs. 17%; p=0.02) 1 5.5 [32]
Retrospective data analysis 825 Heterogeneous subjects from 9 published Phase II and Phase III trials Non-linear relationship (p<0.003) None (analysis not reported) 2b 5b [36]
Descriptive meta-analysis 509 Patients with deep mycosis from 12 reports Seen using graded cut-offs (between 1 and 3 mcg/mL) onlyc Seen with hepato- and neurotoxicity using graded cut-offs (between 3 and 6 mcg/mL) onlyc 1 4 [46]
Prospective cohortd 52 Patients with clinical indications for receiving voriconazole Higher success at > 1 mcg/mL (88% vs. 54%; p=0.02) Seen with encephalopathy > 5.5 mcg/mL (31% vs. 0%; p=0.002) but not with hepatotoxicity 1 5.5 [29]
Population PK analysis 55 Patients with clinical indications for receiving voriconazole Seen with Log-transformed Cmax, Cmin and AUC0–12h and efficacy using oral (p<0.001) but not IV therapy Seen with Cmax, Cmin and AUC0–12h and Grade 3 neurotoxicity (p<0.01) but not hepatotoxicity 1.5 4.5 [43]
Retrospective cohort (multicenter) 201e Patients with clinical indications for receiving voriconazolee Higher success at ≥ 1.7 mcg/mL (93% vs. 74%; p<0.01) Seen with neurotoxicity at > 5 mcg/mL (32% vs. 1.2%; p<0.01) 2 5 [44]
Prospective non-comparative cohort (multicenter) 116f Proven and probable cases of IA Higher success at > 0.25 mcg/mL 6/22 (27%) of patients with plasma concentration > 6 mcg/ml developed abnormal liver function or liver failure 0.25 6 [33]
Retrospective cohort 108g Patients with clinical indications for receiving voriconazole No relationship with clinical or radiological response 12 weeks Levels > 5.5 mcg/mL, not associated with higher rates of encephalopathy or hepatotoxicity NR NR [45]
Retrospective cohort (multicenter) 264h Patients with clinical indications for receiving voriconazole No relationship (higher success when troughs were lower) Very few samples > 5 mcg/mL; incidence of AEs low; underpowered to show exposure-toxicity relationship NR NR [31]
Retrospective data analysis 1053 Subjects from 10 phase II and III trials Not assessed Seen with visual AEs (p=0.01) and AST, ALP and bilirubin (p<0.001) but not ALT (p=0.17) NR NR [47]
Retrospective cohort 26 Patients with IA or invasive candidiasis Not assessed Neurotoxicity hazard ratio 2.27 per 0.1 mcg/mL increase (p< 0.001) NR 4 [48]
Retrospective cohort 39 Patients with clinical indications for receiving voriconazole Not assessed Hepatotoxicity seen with higher initial troughs and sustained levels > 4 mcg/mL NR 4 [50]
Prospective cohort 95 Patients with clinical indications for receiving voriconazole Not assessed Visual hallucinations associated with higher levels (4.5 vs. 2.5 mcg/mL; p=0.004); no relationship seen with hepatotoxicity - NR [49]

TDM= therapeutic drug monitoring; RCT=randomized controlled trial; IFI= invasive fungal infection; AE= adverse event; IA= invasive aspergillosis; AST= aspartate transaminase; ALP= alkaline phosphatase; ALT= alanine transaminase; NR= not reported; Cmax = maximum observed concentration; Cmin = minimum observed concentration; AUC0–12 = area under the concentration versus time curve over the course of a single dosing interval

a

Sub-set of proven or probable IFI

b

trough/MIC ratio

c

relationship not seen when concentration was treated as a continuous variable

d

Cases without TDM (N=39) were studied retrospectively

e

treatment N=170 and prophylaxis N=31

f

116 patients assessed for efficacy and 137 patients assessed for safety

g

Efficacy only evaluated in those with proven or probable IFI (N=46)

h

Efficacy only evaluated in those with proven or probable IA (N=53)

i

Represents studies limited to use of voriconazole with therapeutic intent with a sample size of > 50 for evaluation of efficacy and > 25 when solely for evaluation of toxicity

Voriconazole TDM and efficacy

The only clinical trial investigating the impact of voriconazole TDM on the incidence of adverse drug events and treatment response was a 1:1 randomized, single center study from South Korea. In this study, voriconazole dosage was adjusted using pre-specified algorithms based on trough voriconazole concentrations drawn on day 4 of therapy in the intervention group (55 of 110 patients) in which the targeted range was 1 to 5.5 mcg/mL [32]. The control group received standard voriconazole dosing during the study. Although the incidence of adverse drug events did not differ significantly between groups (p=0.97), the likelihood of voriconazole discontinuation due to adverse effects was 4-fold higher among controls compared to the intervention group (p=0.02). With the availability of voriconazole trough levels to guide dose adjustments, providers tended to continue voriconazole longer in the TDM arm despite the occurrence of a similar number of adverse events. Importantly, TDM was associated with a higher clinical response rate compared to no TDM (81% vs. 59%, p=0.04).

Several observational studies have also evaluated the role of voriconazole TDM, (Table 1), the largest of which involved a secondary analysis of 825 subjects with yeast or mold infections from nine Phase II and Phase III clinical trials. At a mean plasma concentration (Cavg) < 0.5 mcg/mL, the voriconazole response rate was 57% compared to 74% when Cavg was between 0.5 and 5 mcg/mL. Moreover, a nonlinear relationship between Cavg and clinical response was determined by logistic regression (p<0.003) [36]. Higher responses were seen with primary rather than salvage therapy, and with yeast rather than mold infections, and specifically within these groups with Candida spp. rather than Aspergillus spp. Although informative, this study combined patients and pathogens with widely different exposure-response relationships, making it difficult to extrapolate these results to other patient populations.

In a study of voriconazole TDM during 2388 treatment days in 52 patients at a single center investigators found that trough concentrations > 1 mcg/mL yielded a higher response rate (88%) compared to troughs ≤ 1 mcg/mL (54%; p=0.02) [29]. The authors did not find a relationship between voriconazole dose and trough concentration, but did observe that trough concentrations were a significant predictor of clinical response (probability 0.7 at a trough of 1 mcg/mL). In a population pharmacokinetic analysis from 505 plasma concentration values from another 55 patients, the same authors reproduced this concentration-response relationship [43]. In this study, plasma concentrations ranging from 1.5 to 4.5 mcg/mL were associated with a probability of response greater than 85%. The relationship was statistically significant only when voriconazole was administered orally (p<0.001), supporting variability in oral bioavailability as a major determinant of sub-therapeutic voriconazole levels.

Similar findings have been observed in several multi-center studies. In an open-label study involving 201 adult patients, voriconazole trough concentrations were significantly lower (median 0.9 mcg/mL) in patients who failed treatment compared to those who responded (median 2.1 mcg/mL; p<0.05) [44]. Among those patients with proven or probable invasive fungal infections and voriconazole trough concentrations < 1.7 mcg/mL, the treatment failure rate was 35%, compared to a 6% failure rate in patients with voriconazole concentrations ≥ 1.7 mcg/mL. In another multi-center study of 116 assessable patients with invasive aspergillosis, 3 out of 5 patients with voriconazole trough concentrations < 0.25 mcg/mL failed to demonstrate any sustained meaningful treatment response, which underscores the importance of knowing when trough voriconazole concentrations are exceedingly low (< 0.25 mcg/mL) [33]. Several others have found voriconazole TDM to beneficially influence treatment efficacy in studies involving relatively few patients (N < 50) [40,51,52].

In children 2 to 11 years old administered standard adult dosages (3 to 4 mg/kg every 12 h), voriconazole demonstrates linear pharmacokinetics, which has been attributed to higher first-pass metabolism and systemic metabolic rates in the pediatric population [5355]. However, at the recommended dosage for ages 2 to 12 (7 to 8 mg/kg every 12 h), non-linear pharmacokinetics, as seen in adults, are observed [55,56]. The inability to reach adequate levels at standard adult doses (especially in critically ill children [57]) and the variability in trough concentrations at recommended doses, suggests voriconazole TDM may also be helpful in children. Three pediatric studies using voriconazole target trough concentrations ≥ 1 mcg/mL demonstrated a relationship between voriconazole concentrations and efficacy [5860].

Fewer studies have explored the usefulness of voriconazole TDM in the prophylactic setting (i.e. to reduce breakthrough fungal infections) [6164]. Among 93 lung-transplant recipients receiving prophylactic voriconazole, absence of any trough values > 1.5 mcg/mL was associated with a significantly greater number of respiratory cultures growing fungal species (p=0.01) [61]. Notably, some of the positive cultures represented colonization, and the value of preventing fungal colonization remains unclear. A similar study in allogeneic hematopoietic stem cell transplant recipients with hematologic malignancies reported 6 cases of breakthrough candidiasis among 43 patients with voriconazole trough concentrations ≤ 2 mcg/mL and no cases among the 24 patients with concentrations > 2 mcg/mL (p=0.061) [62]. Both of these studies saw four breakthrough infections each with molds [61,62]. Another study in immunocompromised patients failed to show any relationship between TDM of prophylactic voriconazole and efficacy, but involved cases with both prophylactic and therapeutic indications for voriconazole [63]. Current evidence is insufficient to recommend voriconazole target concentrations to ensure adequate prophylaxis.

Some studies failed to demonstrate a relationship between voriconazole monitoring and efficacy [31,41,45,65]. In these studies the number of cases included in the efficacy analysis was low (maximum 53 cases). In one of the studies [41], the trough-efficacy correlation became statistically significant after excluding patients with refractory hematological conditions from the analysis. Another study [45], found a relationship between trough concentrations and efficacy 6, but not 12 weeks after commencing therapy. A meta-analysis of 12 studies was performed to investigate the optimal blood concentration range of voriconazole [46]. Analysis of the extracted data on voriconazole concentrations as a continuous variable from 3 of the 12 studies [41,66,67], and its relationship with efficacy, suggested that the distribution of voriconazole plasma concentrations did not differ significantly between treatment success and failure [Weighted Mean Difference 1.95 (-2.18 to 1.84); p = 0.35]. The addition of one study [29] and dichotomous treatment of extracted data using graded cut-off values between 1 and 3 mcg/mL, demonstrated that a trough of 1 mcg/mL (including a subset analysis limited to cases of invasive aspergillosis), discriminated between treatment success and failure [OR 7.23 (2.84 to 18.37); p<0.0001]. However, the limited number of studies actually analyzed as well as small sample sizes and observational nature, preclude strong conclusions about the drug concentration-efficacy relationship. Notwithstanding these underpowered studies, the majority of studies show a positive benefit of voriconazole TDM on efficacy.

Voriconazole TDM and safety

Voriconazole can produce a variety of adverse effects that vary in severity. Hepatotoxicity, visual disturbances, visual hallucinations, and other neurologic disorders have been directly correlated with plasma concentrations of voriconazole (Table 1). A pooled PK/PD analysis of ten Phase 2 and Phase 3 clinical studies suggested a relationship between voriconazole concentrations and visual disturbances, which occurred in 16% of patients when plasma voriconazole concentrations were < 1 mcg/mL, and rose to 28% at > 9 mcg/mL [47]. Despite this relationship, the usefulness of TDM for this application is limited because voriconazole-associated visual disturbances are typically mild, reversible and generally do not result in discontinuation of therapy [68]. Other neurological adverse effects including visual hallucinations, and less commonly encephalopathy may be more debilitating. Their relationship to voriconazole serum concentrations has been studied as well, with almost all of these studies reporting a statistically significant positive correlation [29,43,44,46,48,49,69].

The association between plasma voriconazole concentrations and hepatotoxicity has been extensively investigated [29,33,41,4347,49,50,70,71]. Investigators reported that 6 of 22 patients with voriconazole concentrations > 6 mcg/mL developed liver function test abnormalities, resulting in 1 death [33]. Based on these and other data TDM was suggested to avoid voriconazole hepatotoxicity [71,72], but this has not been unanimously accepted [73]. The largest PK/PD analysis of voriconazole hepatotoxicityinvestigators [47] reported a statistically significant relationship between voriconazole concentrations and risk of aspartate transaminase, alkaline phosphatase, and bilirubin elevation (p<0.001), but this relationship was not observed for alanine transaminase (p=0.17). More importantly, receiver-operator characteristic curves denoted poor prediction of any liver function tests abnormalities across a range of voriconazole concentrations [47]. One study has suggested that sustained elevated voriconazole concentrations might be associated with an increased risk of hepatotoxicity [50]. Nonetheless, the absolute incidence of hepatotoxicity with voriconazole use remains low, and is comparable to that of other antifungal agents [74]. Thus, there is no universally acceptable concentration threshold, above which voriconazole-related hepatotoxicity is known to occur.

When used in the prophylactic setting, voriconazole toxicity must be weighed against its ability to reduce breakthrough invasive fungal infections. A study evaluating the role of voriconazole in preventing invasive fungal infections among lung transplant recipients demonstrated that toxicities were significant enough to warrant drug discontinuation in 27% (25/93) of cases [61]. The usefulness of voriconazole TDM in the prophylactic setting requires further study.

Voriconazole pharmacokinetics and CYP2C19 polymorphisms

Studies in healthy volunteers

A study in healthy volunteers receiving voriconazole 200 mg or 300 mg po BID x 10 days found that voriconazole Cmax and AUC were increased in CYP2C19 PMs compared to EMs [75]. The impact of CYP2C19 genetic variants on voriconazole pharmacokinetics has been confirmed and characterized in a number studies in adult healthy volunteers (Table 2) [7,8,7680]. Pharmacokinetic parameters including half-life (t1/2) and AUC are significantly increased in CYP2C19 PM compared to CYP2C19 EM receiving oral voriconazole. In single dose studies, the voriconazole t1/2 has ranged from 8.7 hours to 15.2 hours in PM and 3.3 hours to 8.1 hours in EM [7,76,77,79,80]. In a multiple dose study, the voriconazole AUC0-T geometric mean ratio of PM to EM was 3.3 [8]. This increase is consistent with the results of several single dose studies, which determined that the voriconazole AUC0-∞ was 2.8 to 4.1 times higher in PM compared to EM [7,8,7680]. In single oral dose studies significant reductions in voriconazole clearance have been observed in PM versus EM [7,7680]. A multiple dose study reported that the voriconazole apparent oral clearance (Cl/F) decreased from 210 mL/min to 58.3 mL/min in EM compared to PM [8]. However, changes in clearance in this trial were not subjected to statistical evaluation.

Table 2.

Pharmacokinetics of Voriconazole in Healthy Volunteers with CYP2C19 polymorphismsl

Study Design Age
Race
Total N Voriconazole Dosage Regimen CYP2C19 Genotype (N) Pharmacokinetic Parameters Ref.

AUC0-∞ (h x mcg/mL) AUC0-T (h x mcg/mL) Cl (mL/min) t1/2 (h)

RCT, double blinded, placebo controlled, 2 way crossover study (ritonavir/voriconazole DI study) Adults
White
20 Voriconazole 400 mg po x1 CYP2C19*1/*1 (N=8) 16.52 +/−7.21 463 +/−168 8.11 +/−1.35 [76]
CYP2C19*1/*2 (N=8) 22.65 +/−10.88 343 +/−127 8.07 +/−2.22
CYP2C19*2/*2 (N=4) 47.96 +/−23.33a 158 +/−54a 15.21 +/−3.06a

Open label 21 +/−2 yo
Chinese
20 Voriconazole 200 mg po x 1 CYP2C19*1/*17 (N=4) 3.63 +/−0.52b 932.02 +/−120.99b 7.19 +/−3.27 [77]
CYP2C19*1/*1 (N=8) 6.92 +/−2.11 521.53 +/−154.51 8.28 +/−2.02
CYP2C19*2/*2 (N=8) 23.9 +/−5.94c 146.7 +/−33.88c 13.98 +/−3.92c

RCT, open label, 2 way crossover study Adult
White
20 Voriconazole 400 mg po x 1 or voriconazole 400 mg IV x 1
After 14 day washout, voriconazole administered by other route
CYP2C19*1/*1d (N=8) [78]
PO 38.8 +/−12.8e 572 +/−352 7.5 +/−1.2
IV 53.7 +/−22.5e 420 +/−221 7.8 +/−1.4
CYP2C19*1/*2, *1/*3d (N=8)
PO 88.6 +/−40.5e 241 +/−82.1 9.5 +/−5
IV 107 +/−46.7e 194 +/−56 9 +/−4
CYP2C19*2/*2, *2/*3, *3/*3 (N=4)
PO 119 +/−13.8c,e 157 +/−18.8c 9.6 +/−2.3
IV 127 +/−21.7c,e 149 +/−28.3c 9.4 +/−1.6

RCT, open, crossover study (erythromycin/voriconazole DI study) Mean age 19 to 23 yo
Chinese
18 Voriconazole 200 mg po x1 CYP2C19*1/*1 (N=6) 7.7 +/−2.99 499 +/−228 3.72 +/−2.12 [79]
CYP2C19*1/*2 (N=4) 10.75 +/−9.42 452 +/−233 4.09 +/−2.84
CYP2C19*1/*3 (N=2)
CYP2C19*2/*2 (N=5) 30.95 +/−19.2f 153 +/−110g 8.69 +/−5.17g
CYP2C19*2/*3 (N=1)

Analyzed placebo groups of 2 studies (voriconazole and ritonavir or St. John’s Wort) Adults
Caucasian (N=32)
Asian (N=2)
South American (N=1)
35 Voriconazole 400 mg po x 1 CYP2C19*1/*17 (N=8), *2/*17 (N=2) 13.27 +/−3.17 526.9 +/−118 6.96 +/−1.71 [7]
CYP2C19*1/*1 (N=9) 16.44 +/−6.93 465.5 +/−175.7 7.23 +/−2.11
CYP2C19*1/*2 (N=11) 25.66 +/−12.82 319.2 +/−152.2 8.25 +/−3
CYP2C19*2/*2 (N=4) 45.73 +/−20.81a 162.9 +/−47.6a 14.28 +/−3.36a
CYP2C19*2/*2*17(N=1)

RCT, 2 phase crossover study (ginkgo biloba/voriconazole DI study) Adults
Chinese
14 Voriconazole 200 mg po x 1 CYP2C19*1/*1 (N=7) 5.17 (3.73 to 6.88)h 644.85 (484.39 to 894.44)h 3.27 (2.56 to 3.99)h [80]
CYP2C19*2/*2 (N=7) 20.96 (18.81 to 28.45)c,h 159.01 (117.18 to 117.25)c,h 11.26 (10.21 to 16.17)h

Open label parallel group study Adults
Korean
18 Voriconazole 200 mg IV x 1.
1 week wash out.
Voriconazole 200 mg po x 1. Next day voriconazole 200 mg po BID for 5 days, then once on last day
CYP2C19*1/*1 (N=6) [8]
IV 6.513 +/−2.573 563.3 +/−161.7 3.2 +/−2.5
PO (single dose) 5.055 +/−1.771 715 +/−198.3 2.7 +/−1.7
PO (multiple dose) 19.305 +/−9.594 210 +/−108.3 9.6 +/−5.1
CYP2C19*1/*2,*1/*3 (N=6)
IV 10.098 +/−4.151 383.3 +/−160 5.5 +/−3.3
PO (single dose) 8.868 +/−3.644 448.3 +/−221.7 5.8 +/−3.1
PO (multiple dose) 42.369 +/−19.090 98.3 +/−58.3 16.9 +/−7.2
CYP2C19*2/*2, *2/*3 (N=6)
IV 20.479+/−5.077i 171.7 +/−40 13.3 +/−6.1
PO (single dose) 16.582 +/−3.893j 208.3 +/−40 12 +/−3.1
PO (multiple dose) 58.697 +/−11.113k 58.3 +/−15 32.3 +/−9.4

RCT = randomized controlled trial; DI = drug interaction; N = number of patients; AUC0-∞ = area under the concentration versus time curve extrapolated to infinity; AUC0-T = area under the concentration versus time curve from dosing to the time point of the next dose; Cl = clearance; t1/2 = half-life.

a

P<0.01 for *1/*1 versus *2/*2

b

P<0.05 for *1/*1 versus *1/*17

c

P<0.05 for *1/*1 versus *2/*2

d

6 out of 8 EM group were CYPC19*1/*17 and 2 of heterozygous EM group were CYPC19*2/*17 when reanalyzed

e

AUC is AUC0-last in h x nmol/mL

f

P<0.01 for *1/*1 versus *2/*2,*2/*3

g

P<0.05 for *1/*1 versus *2/*2,*2/*3

h

Values are median and interquartile range

i

geometric mean ratio PM/EM 3.23 (90% CI 2.38 to 4.4)

j

geometric mean ratio PM/EM 3.36 (90% CI 2.54 to 4.44)

k

geometric mean ratio PM/EM 3.29 (90% CI 2.22 to 4.9)

l

statistics from studies only included in table for EM versus PM and EM versus URM

Alternations in voriconazole pharmacokinetics in CYP2C19 genetic variants has also been observed with intravenous voriconazole [8,78]. After a single dose (voriconazole 200 mg i.v.), the AUC0-∞ geometric mean ratio of PM to EM was 3.23 in healthy volunteers [8]. In another single dose study (voriconazole 400mg i.v.) the AUC0-∞ was significantly increased and clearance significantly decreased in PM compared to EM [78]. However, in this study 6 out of 8 patients initially categorized as EM were later found to possess the CYP2C19*1/*17 genotype.

Two studies have produced conflicting data on the effect of the CYP2C19*17 allele on voriconazole pharmacokinetics[7,77]. Unfortunately, only volunteers who were heterozygous for the CYP2C19*17 allele (CYP2C19*1/*17) and not homozygous (CYP2C19*17/*17) were enrolled in these trials. One study of healthy volunteers receiving a single oral voriconazole dose (200 mg) demonstrated a significant increase in apparent Cl/F and a decrease in AUC0-∞ in EM (CYPC19*1/*1) compared to URM (CYP2C19*1/*17) [77]. In contrast, an analysis of the placebo groups from two drug interaction studies reported no statistical differences for AUC0-∞, apparent Cl/F, or t1/2 when comparing EM (CYPC19*1/*1) to URM (CYP2C19*1/*17, CYP2C19*2/*17) [7]. Based on the results of these trials, it was proposed that the CYP2C19*17 allele may possibly lead to subtherapeutic voriconazole concentrations [81].

Studies in patients

In contrast to the mostly positive results reported in healthy volunteers, studies on the impact of CYP2C19 genetic variants on voriconazole concentrations in patients have demonstrated conflicting results (Table 3). While retrospective studies have indicated an association between CYP2C19 genotype and voriconazole concentrations [8285], several prospective studies have reported no association [49,86]. For example, a retrospective study in adults reported a minimum observed concentration (Cmin) of 3.67 mg/L and 1.98 mg/L in PM and EM, respectively (P<0.05) [84]. However, in a prospective observational trial in adult Korean patients, median trough concentrations were not significantly different in EM and PM (2.12 mg/L and 2.75 mg/L, respectively p=0.859) [86].

Table 3.

Published Studies on the Relationship of CYP2C19 Polymorphisms on Voriconazole Concentrations and Efficacy and/or Toxicity in Patients.

Study Design Age
Race
Total N Population Voriconazole Dosage Regimen CYP2C19 Genotype (N) CYP2C19 genotype associated with voriconazole: Ref
Concentrations Efficacy Adverse effects
Retrospective study 17 to 84 yo
Race not reported
86 Hematological malignancy Voriconazole 6 mg/kg po BID for 1st 24h, 4 mg/kg po BID days 2 to 7, then 200 mg po BID
12 patients received IV voriconazole for 1st 7 days
CYP2C19 wild typea (N=63)
CYP2C19 non wild typea (N=23)
No significant relationship between CYP2C19 genotype and serum liver enzymes levels [92]
Retrospective review 15 to 40 yo
Caucasian
24 Cystic fibrosis lung transplant recipients Voriconazole 6 mg/kg BID, then MD CYP2C19*1/*17 (N=6)
CYP2C19*17/*17 (N=1)
CYP2C19*1/*1 (N=7)
CYP2C19*1/*2 (N=10)
Voriconazole exposure influenced by CYP2C19 genotype. Proportion of below range concentrations were 37.9%, 15.6%, and 13% in CYP2C19*17, CYP2C19*1, and CYP2C19*2 groups, respectively (P<0.01). No significant relationship between CYP2C19 genotype and adverse drug reactions (P=0.4) [82]
Prospective study 57.3 +/− 19.3 yo
Japanese
29 Fungal infectionsb Voriconazole 6 mg/kg BID for 1 day, then 3.6 +/− 0.8 mg/kg BID
Wild type MD 7.8 +/− 1.9 mg/kg/day and non wild type MD 6.7 +/− 1.2 mg/kg/day
CYP2C19 wild typea (N=10)
CYP2C19 non wild typea (N=19)
No significant relationship between CYP2C19 genotype and hepatotoxicity [70]
Prospective observational trial Adult
Korean
25 Hematological malignancies, probable or proven IA Voriconazole 6 mg/kg IV BID day 1, then 4 mg/kg IV BID
Median MD 7.7 mg/kg/day (interquartile range 7.1 to 8.3)
CYP2C19*1/*1 (N=6)
CYP2C19*1/*2 (N=9)
CYP2C19*1/*3 (N=8)
CYP2C19*2/*2 (N=1)
CYP2C19*3/*3 (N=1)
Trough concentrations did not depend on CYP2C19 genotype
Median trough concentrations in EM, HEM, and PM were 2.12 mg/L, 3.76 mg/L and 2.75 mg/L respectively (p=0.859)
No difference in severe adverse event frequencies between CYP2C19 genotypes [86]
Retrospective study 1 to 15 yo
Japanese
37 Received IV voriconazole and had levels measured Voriconazole median dose 7.7 mg/kg/day (range 3.5 to 18.8 mg/kg/day) CYP2C19*1/*17, 17/*17 (N=2)
CYP2C19*1/*1 (N=12)
CYP2C19*1/*2,*1/*3 (N=16)
CYP2C19*2/*2, *2/*3, *3/*3 (N=7)
Voriconazole concentrations associated with CYP2C19 phenotype. Trough levels higher in PM/HEM group than EM/URM group, median 0.54 mg/dl and 0.06 mg/dl respectively, (P=0.004).
CYP2C19 phenotype not independently associated with high concentrations by multivariate logistic regression analysis
[83]
Prospective observational study 53 +/− 13 yo
Korean
104 Hematologic diseases, probable or proven IA Voriconazole 6 mg/kg IV BID day 1, then 4 mg/kg BID or 200 mg po BID CYP2C19*1/*1 (N=37)
CYP2C19*1/*17c (N=2)
CYP2C19*1/*2 (N=32)
CYP2C19*1/*3 (N=18)
CYP2C19*2/*2 (N=6)
CYP2C19*3/*3 (N=9)
The initial voriconazole trough concentrations 1.8, 2.7, and 3.2 mg/L in EM, HEM, and PM, respectively (P=0.068). No significant difference in treatment response, all cause and IA mortality between CYP2C19 genotypes No significant difference in voriconazole related adverse events between CYP2C19 genotypes [91]
Prospective study 13 to 76 yo
White, non latino (N=59)
White, latino (N=22)
African American (N=16)
Asian (N=3)
95 CYP2C19*1/*1 (N=63)
CYP2C19*1/*2 (N=18)
CYP2C19*1/*3 (N=1)
CYP2C19*2/*2 (N=4)
CYP2C19*1/*9 (N=1)
CYP2C19*1/*11 (N=1)
CYP2C19*1/*15 (N=1)
Other (N=3)
CYP2C19 genotype minor influence over levels. Voriconazole concentration 4.33 mcg/mL and 2.468 mcg/mL in PM and EM, respectively. [49]
Retrospective study 18 to 99 yo
Race not reported
144 Proven or probable invasive fungal infection Dose based on the voriconazole package insert CYP2C19*1/*17 (N=3)
CYP2C19*1/*1 (N=62)
CYP2C19*1/*2,*1/*3 (N=62)
CYP2C19*2/*2,*2/*3 (N=17)
Voriconazole Cmin influenced by CYP2C19 genotype. Cmin higher in PM than EM and HEM (3.67 mg/L, 1.98 mg/L, and 2.36 mg/L, respectively) P<0.05 for EM versus PM [84]
Retrospective review 1 to 19 yo
African (N=6)
European (N=23)
Hispanic (N=2)
Multiple race (N=2)
33 Immunocompromised patients with cancer Initial MD 200 mg BID (≥12 yo), and 7 mg/kg BID (<12 yo)
< 12 yo: dose administered 2.6 to 41.2 mg/kg/day
≥ 12 yo: dose administered 3.6 to 16.1 mg/kg/day
< 12 yo
CYP2C19*17/*17 (N=2)
CYP2C19*1/*17 (N=6)
CYP2C19*1/*1 (N=8)
CYP2C19*1/*2A, *1/*2B (N=3)
≥ 12 yo
CYP2C19*17/*17 (N=2)
CYP2C19*1/*17 (N=2)
CYP2C19*1/*1 (N=3)
CYP2C19*1/*2A, *1/*2B (N=6)
CYP2C19*2A/*2A (N=1)
CYP2C19 diplotype associated with voriconazole concentrations (p=0.002)
Median dose normalized trough concentrations (mcg/mL/mg/kg) 0.01, 0.14, 0.62, 0.07 in CYP2C19*17/*17, CYP2C19*1/*2A, *1/*2B, CYP2C19*2A/*2A, CYP2C19*1/*1 genotype, respectively
[85]

MD = maintenance dose; IA = invasive aspergillosis; Cmin = minimum observed concentration

a

specific genotype not reported in study

b

pulmonary aspergillosis, sinus aspergillosis, pulmonary cryptococcosis, cryptococcal meningitis

c

CYP2C19*1/*17 categorized as EM in this study

The different results in patients compared to healthy volunteers may be due to confounding factors present in patients such as drug interactions, comorbidities and organ dysfunction [87]. Additionally, the CYP2C19 genotype does not appear to account for all of the intrinsic variability in voriconazole pharmacokinetics between individuals. An analysis of placebo groups from two healthy volunteer studies revealed that the CYP2C19 genotype explained 49% and 39% of variability in voriconazole apparent Cl/F and AUC0-∞, respectively [7]. Similarly, in a retrospective review of cystic fibrosis lung transplant recipients, the CYP2C19 genotype only explained 38% of variability in voriconazole maintenance dose [82].

Several studies indicate that the CYP2C19*17 allele may lead to subtherapeutic voriconazole concentrations [82,83,85]. In cystic fibrosis lung transplant recipients, the proportion of below range concentrations was 37.9% and 15.6% in CYP2C19*17 and CYP2C19*1 groups, respectively (P<0.01) [82]. In a retrospective study immunocompromised patients with cancer possessing the CYP2C19*17/*17 genotype had lower median dose-normalized trough concentrations than those with the CYP2C19*1/*1 genotype [85]. In that study, all of the patients with the CYP2C19*17/*17 genotype (N=4) failed to achieve therapeutic voriconazole concentrations. Furthermore, in a retrospective study of patients with voriconazole levels ≤0.2 mcg/mL and excluding those receiving enzyme inducers, allogeneic stem cell transplant or liver transplant patients, inadequate dosing, or timing of levels, the CYP2C19*1/*17 or CYPC219*17/*17 genotypes were found in 8 out of 10 patients (80%) [88]. In complicated dosing settings such as obesity genetic screening may also be informative and clinically helpful. For example, sustained elevations in voriconazole serum concentrations in an obese patient despite appropriate adjusted weight-based dosing were attributed to a CYP2C19 homozygous PM genotype (CYP2C19*2/*2) [89].

Several studies have described voriconazole pharmacokinetics in immunocompromised pediatric patients with CYP2C19 polymorphisms [87,90]. When comparing pediatric PMs to EMs the increase in voriconazole AUC was consistent with the results observed in adults [87,90]. However, the genotypic variability observed in these patients precluded statistical analysis.

Other important issues related to the CYP2C19 genotype include the association between genotype and efficacy or toxicity, and CYP2C19 genotype-guided dosing of voriconazole. Clinical studies have reported that the CYP2C19 genotype is not associated with the efficacy or toxicity of voriconazole (Table 3). Investigators found no significant difference among CYP2C19 genotypes in treatment response, all-cause, and invasive aspergillus mortality [91]. The authors concluded that with therapeutic drug monitoring an association was not observed between CYP2C19 genotype and voriconazole efficacy. Multiple studies have failed to find an association between CYP2C19 genotype and adverse effects [70,82,86,91,92] including hepatotoxicity [70,92]. Nonetheless, several investigators have proposed voriconazole dosing based on CYP2C19 genotype. Matsumoto et al. recommended an initial voriconazole dose of 7.2 to 8.9 mg/kg/day and 4.4 to 6.5 mg/kg/day in CYP2C19 wild type and CYP2C19 non-wild type, respectively in Japanese patients [70]. Wang et al. suggested a voriconazole dose of 200 mg orally or intravenously twice daily in PM and voriconazole 300 mg orally twice daily or 200 mg intravenously twice daily in non-PM [84]. However, to our knowledge a strategy of prospective CYP2C19 genotyping to select an initial voriconazole dose has not been validated prospectively.

Discussion/Conclusion

In this review we describe the evidence supporting the use of voriconazole TDM and the role of CYP2C19 genotyping for voriconazole dosing. The impact of voriconazole TDM on safety and efficacy is still not entirely clear. Most evidence to date was acquired retrospectively in the absence of a non-TDM comparison group and with discretionary post-concentration dosage adjustments. The cost associated with voriconazole TDM is the only identifiable barrier to its routine use. The only randomized controlled trial that assessed the role of voriconazole TDM was from a single-center and underpowered to detect differences in their primary outcome of adverse events [32]. High-volume multicenter randomized controlled trials in this area are currently not available to support definitive guidelines. Until then, we support routine voriconazole TDM given substantial retrospective and prospective observational data supporting its benefit in regards to efficacy, avoidance of neurotoxicity, and minimizing discontinuation of therapy.

While a significant relationship exists in healthy volunteers between CYP2C19 genotype and voriconazole pharmacokinetics, including AUC, Cl/F and t1/2, this association is markedly less apparent in actual patients. Studies also indicate that CYP2C19 genotype is not related to the efficacy or toxicity of voriconazole. Additional studies are needed before routine CYP2C19 genotyping is performed to facilitate initial dose selection of voriconazole. Finally, there is no validated model that allows for an accurate initial dosage of voriconazole based upon CYP2C19 allelic profile. While CYP2C19 genotype data may explain variability of voriconazole serum levels, they alone are not sufficient to guide initial dosing. This is in agreement with several reviews that state further research is needed before the widespread implementation of clinical voriconazole pharmacogenomics [12,14,93,94].

The logistics of timeliness of data from TDM and genotyping remain a continued challenge to patient care. While the “turn around” time for TDM data has improved in both reference laboratories and in hospital laboratories, availability of genotyping data remains difficult. Genotyping data are typically not available for patients as a guide to initial dosing in patients receiving voriconazole therapy.

If the current data are insufficient to recommend CYP2C19 genotyping for all patients, when should it be performed? We feel that institutional resources should first be used to implement voriconazole TDM with a reasonable turn around time. Once voriconazole TDM is implemented, CYP2C19 genotyping, if available, may be a useful adjunct to assist in characterizing the voriconazole disposition of select patients with particularly unpredictable concentrations and a clinical need for aggressive treatment. As a reminder, alternative specimens than blood are needed for CYP2C19 genotyping in allogeneic stem cell or liver transplantation patients and those who have recently received heterologous blood transfusions [95]. We also note that genotyping may be useful in specific clinical situations such as dosing of voriconazole in critically ill patients (especially for patients from ethnicities associated with high rates of PM status such as Asians), in patients on multiple interacting medications, in obese patients receiving intravenous voriconazole, and in selected pediatric patients in whom therapeutic levels are difficult to obtain. In patients receiving initial doses of voriconazole above manufacturer recommendations, CYP2C19 genotyping may be helpful to identify PMs and thereby prevent excessive levels and toxicity. Furthermore, the presence of the CYP2C19*17 allele in critically ill patients may lead to the decision to administer combination antifungal therapy until therapeutic levels of voriconazole are achieved. Knowledge of the CYP2C19 genotype may also aid in the management of voriconazole vincristine drug interactions. In PMs, the half-life of voriconazole may be prolonged, requiring the drug to be held longer than 24 to 48 hours before starting vincristine to avoid this serious drug interaction [96,97]. Similar recommendations were recently made by others assessing of the role of pharmacogenomic screening of patients with hematological malignancies [14].

In conclusion, CYP2C19 genotyping to aid voriconazole dosing is an appealing concept, but further studies are needed before this practice is widely implemented into routine clinical care.

Acknowledgments

This work was supported in part by the intramural research program of the National Institutes of Health. The opinions expressed in this paper are the authors’ and do not reflect those of the National Institutes of Health (NIH) Clinical Center, NIH, Department of Health and Human Services, or the Federal government. Dr. Walsh is a Scholar of the Henry Schueler Foundation and a Scholar of Pediatric Infectious Diseases of the Sharpe Family Foundation. The assistance of Judith Welsh, NIH Library, Office of the Director for the library research is gratefully acknowledged.

References

Papers of special note have been highlighted as:

▪ of interest

  • 1.Vfend (voriconazole) New York, NY: Pfizer; Feb, 2014. Product information. [Google Scholar]
  • 2.Walsh TJ, Anaissie EJ, Denning DW, et al. Treatment of aspergillosis: clinical practice guidelines of the Infectious Diseases Society of America. Clin Infect Dis. 2008;46:327–60. doi: 10.1086/525258. [DOI] [PubMed] [Google Scholar]
  • 3.Pappas PG, Kauffman CA, Andes D, et al. Clinical practice guidelines for the management of candidiasis: 2009 update by the Infectious Diseases Society of America. Clin Infect Dis. 2009;48:503–35. doi: 10.1086/596757. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Theuretzbacher U, Ihle F, Derendorf H. Pharmacokinetic/pharmacodynamic profile of voriconazole. Clin Pharmacokinet. 2006;45:649–63. doi: 10.2165/00003088-200645070-00002. [DOI] [PubMed] [Google Scholar]
  • 5.Andes D, Pascual A, Marchetti O. Antifungal therapeutic drug monitoring: established and emerging indications. Antimicrob Agents Chemother. 2009;53:24–34. doi: 10.1128/AAC.00705-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Geist MJ, Egerer G, Burhenne J, Mikus G. Safety of voriconazole in a patient with CYP2C9*2/CYP2C9*2 genotype. Antimicrob Agents Chemother. 2006;50:3227–8. doi: 10.1128/AAC.00551-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Weiss J, Ten Hoevel MM, Burhenne J, et al. CYP2C19 genotype is a major factor contributing to the highly variable pharmacokinetics of voriconazole. J Clin Pharmacol. 2009;49:196–204. doi: 10.1177/0091270008327537. [DOI] [PubMed] [Google Scholar]
  • 8▪.Lee S, Kim BH, Nam WS, et al. Effect of CYP2C19 polymorphism on the pharmacokinetics of voriconazole after single and multiple doses in healthy volunteers. J Clin Pharmacol. 2012;52:195–203. doi: 10.1177/0091270010395510. A healthy volunteer study in adults that characterized the pharmacokinetics of single (oral and intravenous) and multiple dose (oral) voriconazole in patients with CYP2C19 polymorphisms. [DOI] [PubMed] [Google Scholar]
  • 9.Purkins L, Wood N, Ghahramani P, Greenhalgh K, Allen MJ, Kleinermans D. Pharmacokinetics and safety of voriconazole following intravenous- to oral-dose escalation regimens. Antimicrob Agents Chemother. 2002;46:2546–53. doi: 10.1128/AAC.46.8.2546-2553.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Wang L, McLeod HL, Weinshilboum RM. Genomics and drug response. N Engl J Med. 2011;364:1144–53. doi: 10.1056/NEJMra1010600. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Evans WE, McLeod HL. Pharmacogenomics--drug disposition, drug targets, and side effects. N Engl J Med. 2003;348:538–49. doi: 10.1056/NEJMra020526. [DOI] [PubMed] [Google Scholar]
  • 12▪.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;34:703–18. doi: 10.1002/phar.1400. A recent and extensive review describing the pharmacogenomics of voriconazole. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. [accessed, February 4, 2015]; http://www.pharmgkb.org/drug/PA10233#tabview=tab0&subtab=31.
  • 14.Chau MM, Kong DC, van Hal SJ, et al. Consensus guidelines for optimizing antifungal drug delivery and monitoring to avoid toxicity and improve outcomes in patients with haematological malignancy, 2014. Intern Med J. 2014;44:1364–88. doi: 10.1111/imj.12600. [DOI] [PubMed] [Google Scholar]
  • 15.Desta Z, Zhao X, Shin JG, Flockhart DA. Clinical significance of the cytochrome P450 2C19 genetic polymorphism. Clin Pharmacokinet. 2002;41:913–58. doi: 10.2165/00003088-200241120-00002. [DOI] [PubMed] [Google Scholar]
  • 16.Ingelman-Sundberg M. Pharmacogenetics of cytochrome P450 and its applications in drug therapy: the past, present and future. Trends Pharmacol Sci. 2004;25:193–200. doi: 10.1016/j.tips.2004.02.007. [DOI] [PubMed] [Google Scholar]
  • 17.Hirota T, Eguchi, Ieri I. Impact of genetic polymorphisms in CYP2C9 and CYP2C19 on the pharmacokinetics of clinically used drugs. Drug Metab Pharmacokinet. 2013;28:28–37. doi: 10.2133/dmpk.dmpk-12-rv-085. [DOI] [PubMed] [Google Scholar]
  • 18. [accessed, January 10, 2015]; http://www.cypalleles.ki.se/cyp2c19.htm.
  • 19.Frye RF. In: Pharmacogenetics of oxidative drug metabolism and its clinical applications in Pharmacogenomics: applications to patient care. McLeod HL, DeVane CL, Haga SB, et al., editors. American College of Clinical Pharmacy; Lenexa, Kansas: 2009. pp. 32–53. [Google Scholar]
  • 20.Bertisson L. Geographical/interracial differences in polymorphic drug oxidation. Current state of knowledge of cytochromes P450 (CYP) 2D6 and 2C19. Clin Pharmacokinet. 1995;29:192–209. doi: 10.2165/00003088-199529030-00005. [DOI] [PubMed] [Google Scholar]
  • 21.Scott SA, Sangkuhl K, Stein CM, et al. Clinical Pharmacogenetics Implementation Consortium guidelines for CYP2C19 genotype and clopidogrel therapy: 2013 update. Clin Pharmacol Ther. 2013;94:317–23. doi: 10.1038/clpt.2013.105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Strom CM, Goos D, Crossley B, et al. Testing for variants in CYP2C19: population frequencies and testing experience in a clinical laboratory. Genet Med. 2012;14:95–100. doi: 10.1038/gim.0b013e3182329870. [DOI] [PubMed] [Google Scholar]
  • 23.Sugimoto K, Uno T, Yamazaki H, Tateishi T. Limited frequency of the CYP2C19*17 allele and its minor role in a Japanese population. Br J Clin Pharmacol. 2008;65:437–9. doi: 10.1111/j.1365-2125.2007.03057.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Andes D, Lepak A. Editorial commentary: Antifungal therapeutic drug monitoring progress: getting it right the first time. Clin Infect Dis. 2012;55:391–3. doi: 10.1093/cid/cis446. [DOI] [PubMed] [Google Scholar]
  • 25.Sim SC, Risinger C, Dahl ML, et al. A common novel CYP2C19 gene variant causes ultrarapid drug metabolism relevant for the drug response to proton pump inhibitors and antidepressants. Clin Pharmacol Ther. 2006;79:103–113. doi: 10.1016/j.clpt.2005.10.002. [DOI] [PubMed] [Google Scholar]
  • 26.Johnston A. The pharmacokinetics of voriconazole. Br J Clin Pharmacol. 2003;56 (Suppl 1):1. doi: 10.1046/j.1365-2125.2003.01991.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Trifilio S, Pennick G, Pi J, et al. Monitoring plasma voriconazole levels may be necessary to avoid subtherapeutic levels in hematopoietic stem cell transplant recipients. Cancer. 2007;109:1532–5. doi: 10.1002/cncr.22568. [DOI] [PubMed] [Google Scholar]
  • 28.Bruggemann RJ, Donnelly JP, Aarnoutse RE, et al. Therapeutic drug monitoring of voriconazole. Ther Drug Monit. 2008;30:403–11. doi: 10.1097/FTD.0b013e31817b1a95. [DOI] [PubMed] [Google Scholar]
  • 29.Pascual A, Calandra T, Bolay S, Buclin T, Bille J, Marchetti O. Voriconazole therapeutic drug monitoring in patients with invasive mycoses improves efficacy and safety outcomes. Clin Infect Dis. 2008;46:201–11. doi: 10.1086/524669. [DOI] [PubMed] [Google Scholar]
  • 30.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;53:1793–6. doi: 10.1128/AAC.01316-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Racil Z, Winterova J, Kouba M, et al. Monitoring trough voriconazole plasma concentrations in haematological patients: real life multicentre experience. Mycoses. 2012;55:483–92. doi: 10.1111/j.1439-0507.2012.02186.x. [DOI] [PubMed] [Google Scholar]
  • 32▪.Park WB, Kim NH, Kim KH, et al. The effect of therapeutic drug monitoring on safety and efficacy of voriconazole in invasive fungal infections: a randomized controlled trial. Clin Infect Dis. 2012;55:1080–7. doi: 10.1093/cid/cis599. Randomized, assessor-blinded, controlled, single center trial evaluating clinical utility of therapeutic drug monitoring for voriconazole in invasive fungal infections found reduced drug discontinuation due to adverse events, and improved treatment response. [DOI] [PubMed] [Google Scholar]
  • 33.Denning DW, Ribaud P, Milpied N, et al. Efficacy and safety of voriconazole in the treatment of acute invasive aspergillosis. Clin Infect Dis. 2002;34:563–71. doi: 10.1086/324620. [DOI] [PubMed] [Google Scholar]
  • 34.Goodwin ML, Drew RH. Antifungal serum concentration monitoring: an update. J Antimicrob Chemother. 2008;61:17–25. doi: 10.1093/jac/dkm389. [DOI] [PubMed] [Google Scholar]
  • 35.Andes D, Marchillo K, Stamstad T, Conklin R. In vivo pharmacokinetics and pharmacodynamics of a new triazole, voriconazole, in a murine candidiasis model. Antimicrob Agents Chemother. 2003;47:3165–9. doi: 10.1128/AAC.47.10.3165-3169.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Troke PF, Hockey HP, Hope WW. Observational study of the clinical efficacy of voriconazole and its relationship to plasma concentrations in patients. Antimicrob Agents Chemother. 2011;55:4782–8. doi: 10.1128/AAC.01083-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Howard A, Hoffman J, Sheth A. Clinical application of voriconazole concentrations in the treatment of invasive aspergillosis. Ann Pharmacother. 2008;42:1859–64. doi: 10.1345/aph.1L243. [DOI] [PubMed] [Google Scholar]
  • 38.Johnson LB, Kauffman CA. Voriconazole: a new triazole antifungal agent. Clin Infect Dis. 2003;36:630–7. doi: 10.1086/367933. [DOI] [PubMed] [Google Scholar]
  • 39.Pfaller MA, Diekema DJ, Rex JH, et al. Correlation of MIC with outcome for Candida species tested against voriconazole: analysis and proposal for interpretive breakpoints. J Clin Microbiol. 2006;44:819–26. doi: 10.1128/JCM.44.3.819-826.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Smith J, Safdar N, Knasinski V, et al. Voriconazole therapeutic drug monitoring. Antimicrob Agents Chemother. 2006;50:1570–2. doi: 10.1128/AAC.50.4.1570-1572.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Ueda K, Nannya Y, Kumano K, et al. Monitoring trough concentration of voriconazole is important to ensure successful antifungal therapy and to avoid hepatic damage in patients with hematological disorders. Int J Hematol. 2009;89:592–9. doi: 10.1007/s12185-009-0296-3. [DOI] [PubMed] [Google Scholar]
  • 42.Davies-Vorbrodt S, Ito JI, Tegtmeier BR, Dadwal SS, Kriengkauykiat J. Voriconazole serum concentrations in obese and overweight immunocompromised patients: a retrospective review. Pharmacotherapy. 2013;33:22–30. doi: 10.1002/phar.1156. [DOI] [PubMed] [Google Scholar]
  • 43▪.Pascual A, Csajka C, Buclin T, et al. 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;55:381–90. doi: 10.1093/cid/cis437. A population-pharmacokinetics analysis involving 55 patients with invasive mycoses suggested that a therapeutic range for voriconazole plasma concentrations between 1.5 mg/L and 4.5 mg/L provided a > 85% probability of response and < 15% probability of neurotoxicity. [DOI] [PubMed] [Google Scholar]
  • 44▪.Dolton MJ, Ray JE, Chen SC, Ng K, Pont LG, McLachlan AJ. Multicenter study of voriconazole pharmacokinetics and therapeutic drug monitoring. Antimicrob Agents Chemother. 2012;56:4793–9. doi: 10.1128/AAC.00626-12. In a multicenter retrospective study of the relationships between voriconazole concentrations and clinical outcomes and adverse events in 201 patients, treatment success was significantly greater at voriconazole concentrations ≥1.7 mcg/mL (p<0.01) and neurotoxicity occurred more frequently at concentrations > 5 mcg/mL (p<0.01) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Chu HY, Jain R, Xie H, Pottinger P, Fredricks DN. Voriconazole therapeutic drug monitoring: retrospective cohort study of the relationship to clinical outcomes and adverse events. BMC Infect Dis. 2013;13:105. doi: 10.1186/1471-2334-13-105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Hamada Y, Seto Y, Yago K, Kuroyama M. Investigation and threshold of optimum blood concentration of voriconazole: a descriptive statistical meta-analysis. J Infect Chemother. 2012;18:501–7. doi: 10.1007/s10156-011-0363-6. [DOI] [PubMed] [Google Scholar]
  • 47.Tan K, Brayshaw N, Tomaszewski K, Troke P, Wood N. Investigation of the potential relationships between plasma voriconazole concentrations and visual adverse events or liver function test abnormalities. J Clin Pharmacol. 2006;46:235–43. doi: 10.1177/0091270005283837. [DOI] [PubMed] [Google Scholar]
  • 48.Imhof A, Schaer DJ, Schanz U, Schwarz U. Neurological adverse events to voriconazole: evidence for therapeutic drug monitoring. Swiss Med Wkly. 2006;136:739–42. doi: 10.4414/smw.2006.11547. [DOI] [PubMed] [Google Scholar]
  • 49.Zonios D, Yamazaki H, Murayama N, et al. Voriconazole metabolism, toxicity, and the effect of cytochrome P450 2C19 genotype. J Infect Dis. 2014;209:1941–8. doi: 10.1093/infdis/jiu017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Suzuki Y, Tokimatsu I, Sato Y, et al. Association of sustained high plasma trough concentration of voriconazole with the incidence of hepatotoxicity. Clin Chim Acta. 2013;424:119–22. doi: 10.1016/j.cca.2013.05.025. [DOI] [PubMed] [Google Scholar]
  • 51.Miyakis S, van Hal SJ, Ray J, Marriott D. Voriconazole concentrations and outcome of invasive fungal infections. Clin Microbiol Infect. 2010;16:927–33. doi: 10.1111/j.1469-0691.2009.02990.x. [DOI] [PubMed] [Google Scholar]
  • 52.Gomez-Lopez A, Cendejas-Bueno E, Cuesta I, et al. Voriconazole serum levels measured by high-performance liquid chromatography: a monocentric study in treated patients. Med Mycol. 2012;50:439–45. doi: 10.3109/13693786.2011.630039. [DOI] [PubMed] [Google Scholar]
  • 53.Walsh TJ, Karlsson MO, Driscoll T, et al. Pharmacokinetics and safety of intravenous voriconazole in children after single- or multiple-dose administration. Antimicrob Agents Chemother. 2004;48:2166–72. doi: 10.1128/AAC.48.6.2166-2172.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Chen J, Chan C, Colantonio D, Seto W. Therapeutic drug monitoring of voriconazole in children. Ther Drug Monit. 2012;34:77–84. doi: 10.1097/FTD.0b013e31823f3516. [DOI] [PubMed] [Google Scholar]
  • 55.Karlsson MO, Lutsar I, Milligan PA. Population pharmacokinetic analysis of voriconazole plasma concentration data from pediatric studies. Antimicrob Agents Chemother. 2009;53:935–44. doi: 10.1128/AAC.00751-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Walsh TJ, Driscoll T, Milligan PA, et al. Pharmacokinetics, safety, and tolerability of voriconazole in immunocompromised children. Antimicrob Agents Chemother. 2010;54:4116–23. doi: 10.1128/AAC.00896-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Alffenaar JW, Doedens RA, Groninger E, Kosterink JG. High-dose voriconazole in a critically ill pediatric patient with neuroblastoma. Pediatr Infect Dis J. 2008;27:189–90. doi: 10.1097/INF.0b013e31815c89fd. [DOI] [PubMed] [Google Scholar]
  • 58.Neely M, Rushing T, Kovacs A, Jelliffe R, Hoffman J. Voriconazole pharmacokinetics and pharmacodynamics in children. Clin Infect Dis. 2010;50:27–36. doi: 10.1086/648679. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Choi SH, Lee SY, Hwang JY, et al. Importance of voriconazole therapeutic drug monitoring in pediatric cancer patients with invasive aspergillosis. Pediatr Blood Cancer. 2013;60:82–7. doi: 10.1002/pbc.24262. [DOI] [PubMed] [Google Scholar]
  • 60.Soler-Palacin P, Frick MA, Martin-Nalda A, et al. Voriconazole drug monitoring in the management of invasive fungal infection in immunocompromised children: a prospective study. J Antimicrob Chemother. 2012;67:700–6. doi: 10.1093/jac/dkr517. [DOI] [PubMed] [Google Scholar]
  • 61.Mitsani D, Nguyen MH, Shields RK, et al. Prospective, observational study of voriconazole therapeutic drug monitoring among lung transplant recipients receiving prophylaxis: factors impacting levels of and associations between serum troughs, efficacy, and toxicity. Antimicrob Agents Chemother. 2012;56:2371–7. doi: 10.1128/AAC.05219-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Trifilio S, Singhal S, Williams S, et al. Breakthrough fungal infections after allogeneic hematopoietic stem cell transplantation in patients on prophylactic voriconazole. Bone Marrow Transplant. 2007;40:451–6. doi: 10.1038/sj.bmt.1705754. [DOI] [PubMed] [Google Scholar]
  • 63.Pieper S, Kolve H, Gumbinger HG, Goletz G, Wurthwein G, Groll AH. Monitoring of voriconazole plasma concentrations in immunocompromised paediatric patients. J Antimicrob Chemother. 2012;67:2717–24. doi: 10.1093/jac/dks258. [DOI] [PubMed] [Google Scholar]
  • 64.Barreto JN, Beach CL, Wolf RC, et al. The incidence of invasive fungal infections in neutropenic patients with acute leukemia and myelodysplastic syndromes receiving primary antifungal prophylaxis with voriconazole. Am J Hematol. 2013;88:283–8. doi: 10.1002/ajh.23388. [DOI] [PubMed] [Google Scholar]
  • 65.Lee YJ, Lee SO, Choi SH, et al. Initial voriconazole trough blood levels and clinical outcomes of invasive aspergillosis in patients with hematologic malignancies. Med Mycol. 2013;51:324–30. doi: 10.3109/13693786.2012.694082. [DOI] [PubMed] [Google Scholar]
  • 66.Hagiwara E, Shiihara J, Matsushima A, et al. Usefulness of monitoring plasma voriconazole concentration in patients with chronic necrotizing pulmonary aspergillosis. Nihon Kokyuki Gakkai Zasshi. 2009;47:93–7. [PubMed] [Google Scholar]
  • 67.Okuda T, Okuda A, Watanabe N, Takao M, Takayanagi K. Retrospective serological tests for determining the optimal blood concentration of voriconazole for treating fungal infection. Yakugaku Zasshi. 2008;128:1811–8. doi: 10.1248/yakushi.128.1811. [DOI] [PubMed] [Google Scholar]
  • 68.Boucher HW, Groll AH, Chiou CC, Walsh TJ. Newer systemic antifungal agents: pharmacokinetics, safety and efficacy. Drugs. 2004;64:1997–2020. doi: 10.2165/00003495-200464180-00001. [DOI] [PubMed] [Google Scholar]
  • 69.Zonios DI, Gea-Banacloche J, Childs R, Bennett JE. Hallucinations during voriconazole therapy. Clin Infect Dis. 2008;47:e7–e10. doi: 10.1086/588844. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Matsumoto K, Ikawa K, Abematsu K, et al. Correlation between voriconazole trough plasma concentration and hepatotoxicity in patients with different CYP2C19 genotypes. Int J Antimicrob Agents. 2009;34:91–4. doi: 10.1016/j.ijantimicag.2009.01.008. [DOI] [PubMed] [Google Scholar]
  • 71.Trifilio S, Ortiz R, Pennick G, et al. Voriconazole therapeutic drug monitoring in allogeneic hematopoietic stem cell transplant recipients. Bone Marrow Transplant. 2005;35:509–13. doi: 10.1038/sj.bmt.1704828. [DOI] [PubMed] [Google Scholar]
  • 72.Potoski BA, Brown J. The safety of voriconazole. Clin Infect Dis. 2002;35:1273–5. doi: 10.1086/343746. [DOI] [PubMed] [Google Scholar]
  • 73.Lutsar I, Hodges MR, Tomaszewski K, Troke PF, Wood ND. Safety of voriconazole and dose individualization. Clin Infect Dis. 2003;36:1087–8. doi: 10.1086/374248. [DOI] [PubMed] [Google Scholar]
  • 74.Walsh TJ, Pappas P, Winston DJ, et al. Voriconazole compared with liposomal amphotericin B for empirical antifungal therapy in patients with neutropenia and persistent fever. N Engl J Med. 2002;346:225–34. doi: 10.1056/NEJM200201243460403. [DOI] [PubMed] [Google Scholar]
  • 75.Ikeda Y, Umemura K, Kondo K, Sekiguchi K, Miyoshi S, Nakashima M. Pharmacokinetics of voriconazole and cytochrome P450 2C19 genetic status. Clin Pharmacol Ther. 2004;75:587–8. doi: 10.1016/j.clpt.2004.02.002. [DOI] [PubMed] [Google Scholar]
  • 76.Mikus G, Schöwel V, Drzewinska M, et al. Potent cytochrome P450 2C19 genotype-related interaction between voriconazole and the cytochrome P450 3A4 inhibitor ritonavir. Clin Pharmacol Ther. 2006;80:126–35. doi: 10.1016/j.clpt.2006.04.004. [DOI] [PubMed] [Google Scholar]
  • 77.Wang G, Lei HP, Li Z, et al. The CYP2C19 ultra-rapid metabolizer genotype influences the pharmacokinetics of voriconazole in healthy male volunteers. Eur J Clin Pharmacol. 2009;65:281–5. doi: 10.1007/s00228-008-0574-7. [DOI] [PubMed] [Google Scholar]
  • 78.Scholz I, Oberwittler H, Riedel KD, et al. Pharmacokinetics, metabolism and bioavailability of the triazole antifungal agent voriconazole in relation to CYP2C19 genotype. Br J Clin Pharmacol. 2009;68:906–15. doi: 10.1111/j.1365-2125.2009.03534.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Shi HY, Yan J, Zhu WH, et al. Effects of erythromycin on voriconazole pharmacokinetics and association with CYP2C19 polymorphism. Eur J Clin Pharmacol. 2010;66:1131–6. doi: 10.1007/s00228-010-0869-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Lei HP, Wang G, Wang LS, et al. Lack of effect of Ginkgo biloba on voriconazole pharmacokinetics in Chinese volunteers identified as CYP2C19 poor and extensive metabolizers. Ann Pharmacother. 2009;43:726–31. doi: 10.1345/aph.1L537. [DOI] [PubMed] [Google Scholar]
  • 81.Dolton MJ, McLachlan AJ. Clinical importance of the CYP2C19*17 variant allele for voriconazole. Br J Clin Pharmacol. 2011;71:137–8. doi: 10.1111/j.1365-2125.2010.03801.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Berge M, Guillemain R, Trégouet DA, et al. Effect of cytochrome P450 2C19 genotype on voriconazole exposure in cystic fibrosis lung transplant patients. Eur J Clin Pharmacol. 2011;67:253–60. doi: 10.1007/s00228-010-0914-2. [DOI] [PubMed] [Google Scholar]
  • 83.Narita A, Muramatsu H, Sakaguchi H, et al. Correlation of CYP2C19 phenotype with voriconazole plasma concentration in children. J Pediatr Hematol Oncol. 2013;35:e219–23. doi: 10.1097/MPH.0b013e3182880eaa. [DOI] [PubMed] [Google Scholar]
  • 84.Wang T, Zhu H, Sun J, et al. Efficacy and safety of voriconazole and CYP2C19 polymorphism for optimised dosage regimens in patients with invasive fungal infections. Int J Antimicrob Agents. 2014;44:436–42. doi: 10.1016/j.ijantimicag.2014.07.013. [DOI] [PubMed] [Google Scholar]
  • 85.Hicks JK, Crews KR, Flynn P, et al. Voriconazole plasma concentrations in immunocompromised pediatric patients vary by CYP2C19 diplotypes. Pharmacogenomics. 2014;15:1065–78. doi: 10.2217/pgs.14.53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Kim SH, Yim DS, Choi SM, et al. Voriconazole-related severe adverse events: clinical application of therapeutic drug monitoring in Korean patients. Int J Infect Dis. 2011;15:e753–8. doi: 10.1016/j.ijid.2011.06.004. [DOI] [PubMed] [Google Scholar]
  • 87.Driscoll TA, Frangoul H, Nemecek ER, et al. Comparison of pharmacokinetics and safety of voriconazole intravenous-to-oral switch in immunocompromised adolescents and healthy adults. Antimicrob Agents Chemother. 2011;55:5780–9. doi: 10.1128/AAC.05010-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Hassan A, Burhenne J, Riedel KD, et al. Modulators of very low voriconazole concentrations in routine therapeutic drug monitoring. Ther Drug Monit. 2011;33:86–93. doi: 10.1097/FTD.0b013e31820530cd. [DOI] [PubMed] [Google Scholar]
  • 89.Moriyama B, Jarosinski P, Figg WD, et al. Pharmacokinetics of intravenous voriconazole in obese patients: implications of CYP2C19 homozygous poor metabolizer genotype. Pharmacotherapy. 2013;33:e19–22. doi: 10.1002/phar.1192. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Mori M, Kobayashi R, Kato K, et al. Pharmacokinetics and safety of voriconazole intravenous-to-oral switch regimens in immunocompromised japanese pediatric patients. Antimicrob Agents Chemother. 2015;59:1004–13. doi: 10.1128/AAC.04093-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91▪.Kim SH, Lee DG, Kwon JC, et al. Clinical Impact of Cytochrome P450 2C19 Genotype on the Treatment of Invasive Aspergillosis under Routine Therapeutic Drug Monitoring of Voriconazole in a Korean Population. Infect Chemother. 2013;45:406–14. doi: 10.3947/ic.2013.45.4.406. A prospective observational study found with routine therapeutic drug monitoring there was no significant relationship between CYP2C19 genotype and outcome or toxicity with voriconazole in patients with invasive aspergillosis. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Levin MD, den Hollander JG, van der Holt B, et al. Hepatotoxicity of oral and intravenous voriconazole in relation to cytochrome P450 polymorphisms. J Antimicrob Chemother. 2007;60:1104–7. doi: 10.1093/jac/dkm330. [DOI] [PubMed] [Google Scholar]
  • 93.Dolton MJ, McLachlan AJ. Optimizing azole antifungal therapy in the prophylaxis and treatment of fungal infections. Curr Opin Infect Dis. 2014;27:493–500. doi: 10.1097/QCO.0000000000000103. [DOI] [PubMed] [Google Scholar]
  • 94.Dolton MJ, McLachlan AJ. Voriconazole pharmacokinetics and exposure-response relationships: assessing the links between exposure, efficacy and toxicity. Int J Antimicrob Agents. 2014;44:183–93. doi: 10.1016/j.ijantimicag.2014.05.019. [DOI] [PubMed] [Google Scholar]
  • 95. [assessed, January 29, 2015]; http://www.mayomedicallaboratories.com/test-catalog/Clinical+and+Interpretive/62996.
  • 96.Moriyama B, Henning SA, Leung J, et al. Adverse interactions between antifungal azoles and vincristine: review and analysis of cases. Mycoses. 2012;55:290–7. doi: 10.1111/j.1439-0507.2011.02158.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Moriyama B, Falade-Nwulia O, Leung J, et al. Prolonged half-life of voriconazole in a CYP2C19 homozygous poor metabolizer receiving vincristine chemotherapy: avoiding a serious adverse drug interaction. Mycoses. 2011;54:e877–9. doi: 10.1111/j.1439-0507.2011.02016.x. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES