You are on page 1of 8

International Journal of Infectious Diseases 93 (2020) 345–352

Contents lists available at ScienceDirect

International Journal of Infectious Diseases


journal homepage: www.elsevier.com/locate/ijid

Pharmacokinetics of intravenous voriconazole in patients with


liver dysfunction: A prospective study in the intensive care unit$
Xiao-bin Lina,1, Fa Huangb,1, Li Tongb , Yan-zhe Xiaa , Jing-jing Wua , Jia Lia ,
Xiao-guang Hub , Tao Liangc, Xiao-man Liua , Guo-ping Zhongd, Chang-jie Caib,** ,
Xiao Chena,*
a
Department of Pharmacy, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
b
Department of Critical Care Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
c
School of Pharmacy, Xinhua College of Sun Yat-sen University, Guangzhou 510520, China
d
Institute of Clinical Pharmacology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510080, China

A R T I C L E I N F O A B S T R A C T

Article history: Objectives: To characterize the pharmacokinetics (PK) of intravenous voriconazole (VRC) in critically ill
Received 29 November 2019 patients with liver dysfunction.
Received in revised form 18 February 2020 Methods: Patients with liver dysfunction in the intensive care unit (ICU) were included prospectively. The
Accepted 19 February 2020
Child–Pugh score was used to categorize the degree of liver dysfunction. The initial intravenous VRC
dosing regimen comprised a loading dose of 300 mg every 12 h for the first 24 h, followed by 200 mg
Keywords: every 12 h. The first PK curves (PK curve 1) were drawn within one dosing interval of the first dose for 17
Voriconazole
patients; the second PK curves (PK curve 2) were drawn within one dosing interval after a minimum of
Liver dysfunction
Pharmacokinetics
seven doses for 12 patients. PK parameters were estimated by non-compartmental analysis.
Child–Pugh score Results: There were good correlations between the area under the curve (AUC0–12) of PK curve 2 and the
corresponding trough concentration (C0) and peak concentration (Cmax) (r2 = 0.951 and 0.963,
respectively; both p < 0.001). The median half-life (t1/2) and clearance (CL) of patients in Child–Pugh class
A (n = 3), B (n = 5), and C (n = 4) of PK curve 2 were 24.4 h and 3.31 l/h, 29.1 h and 2.54 l/h, and 60.7 h and
2.04 l/h, respectively. In the different Child–Pugh classes, the CL (median) of PK curve 2 were all lower
than those of PK curve 1. The apparent steady-state volume of distribution (Vss) of PK curve 1 was
positively correlated with actual body weight (r2 = 0.450, p = 0.004). The median first C0 of 17 patients
determined on day 5 was 5.27 (2.61) mg/ml, and 29.4% of C0 exceeded the upper limit of the therapeutic
window (2–6 mg/ml).
Conclusions: The CL of VRC decreased with increasing severity of liver dysfunction according to the Child–
Pugh classification, along with an increased t1/2, which resulted in high plasma exposure of VRC. Adjusted
dosing regimens of intravenous VRC should be established based on Child–Pugh classes for these ICU
patients, and plasma concentrations should be monitored closely to avoid serious adverse events.
© 2020 The Authors. Published by Elsevier Ltd on behalf of International Society for Infectious Diseases.
This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-
nd/4.0/).

$
This clinical study is registered in the Chinese Clinical Trial Registry (http://
Introduction
www.chictr.org.cn, registration number ChiCTR1800019472).
* Corresponding author at: Department of Pharmacy, The First Affiliated Hospital
of Sun Yat-sen University, No. 58, Zhongshan 2nd Road, Guangzhou 510080, China. Patients with liver dysfunction, especially those in the intensive
** Corresponding author at: Department of Critical Care Medicine, The First care unit (ICU), become susceptible to fungi because of decreased
Affiliated Hospital of Sun Yat-sen University, No. 58, Zhongshan 2nd Road,
immune function and increased intestinal permeability (Koulenti
Guangzhou 510080, China.
E-mail addresses: linxb6@mail2.sysu.edu.cn (X.-b. Lin), et al., 2014; Yamada et al., 2018). The mortality of invasive
huangf56@mail2.sysu.edu.cn (F. Huang), tongli2@mail.sysu.edu.cn (L. Tong), aspergillosis in these patients is extremely high (Chen et al., 2013;
sumtreexyz@163.com (Y.-z. Xia), wujingjingsysu@163.com (J.-j. Wu), Jeurissen et al., 2013). Voriconazole (VRC), a second-generation
pharma_jiali@163.com (J. Li), huxiaog@mail.sysu.edu.cn (X.-g. Hu), triazole with potent broad-spectrum antifungal activity, is
ltao_work@163.com (T. Liang), 56873620@qq.com (X.-m. Liu),
zhonggp@mail.sysu.edu.cn (G.-p. Zhong), caichjie@mail.sysu.edu.cn (C.-j. Cai),
recommended as a first-line agent for the treatment of invasive
chenx5@mail.sysu.edu.cn (X. Chen). aspergillosis (Patterson et al., 2016) and as an alternative therapy
1
Xiao-bin Lin and Fa Huang contributed equally to this work. for candidemia (Pappas et al., 2016).

https://doi.org/10.1016/j.ijid.2020.02.041
1201-9712/© 2020 The Authors. Published by Elsevier Ltd on behalf of International Society for Infectious Diseases. This is an open access article under the CC BY-NC-ND
license (http://creativecommons.org/licenses/by-nc-nd/4.0/).
346 X.- Lin et al. / International Journal of Infectious Diseases 93 (2020) 345–352

VRC is metabolized in the liver principally by the cytochrome mg every 12 h for the first 24 h, followed by a maintenance dose
P450 (CYP) 2C19 isoenzyme and, to a lesser extent, by CYP3A4 and of 200 mg every 12 h. A PK curve (PK curve 1) was drawn at t = 0
CYP2C9 (Hyland et al., 2003). A growing number of studies have (pre-dose), 1, 2, 4, 6, 8, and 12 h from the start of the first VRC
shown that CYP2C19 polymorphisms make a crucial contribution infusion; a second PK curve (PK curve 2) was drawn at similar
to the extensive pharmacokinetic (PK) variability of VRC (Lee et al., sampling time-points after the administration of at least seven
2012; Li et al., 2017; Lin et al., 2018; Wang et al., 2014a). doses. Additional C0 samples were collected within 30 minutes
Additionally, age, liver function, co-medications, and inflammation before the next dose based on routine clinical TDM.
could also contribute to PK variability (Dolton et al., 2014;
Theuretzbacher et al., 2006; Veringa et al., 2017; Wang et al., Data collection
2014a). The efficacy and toxicity of VRC are considered to be
significantly correlated with plasma trough concentrations (C0), A standardized data collection form was used to individually
and the narrow therapeutic range has been identified. Therefore, review the medical records of the patients. Researchers closely
therapeutic drug monitoring (TDM) of VRC is widely recom- recorded the dosing information (e.g., VRC dose, administration
mended in clinical practice to maximize efficacy and minimize time, duration of infusion, and sampling time), co-medications
toxicity (Dolton and McLachlan, 2014). such as proton pump inhibitors (PPIs), and biochemical indices
The liver plays an important role in the hepatic metabolism or starting at patient enrolment. In addition, demographic data (age,
biliary excretion of the majority of medicines, and it may affect sex, and actual body weight) and disease scores (Acute Physiology
plasma protein binding, thus affecting the distribution and and Chronic Health Evaluation (APACHE) II score within 24 h of ICU
elimination of drugs (Verbeeck, 2008). One study (Alffenaar admission, sequential organ failure assessment (SOFA) score at
et al., 2009) reported that decreased hepatic function could baseline, and Child–Pugh score) were collected.
significantly reduce VRC metabolism, resulting in high plasma VRC
levels. Recently, supratherapeutic C0 of VRC under empirical Analytical assays
treatment regimens were observed in patients with moderate and
serious hepatic cirrhosis (Child–Pugh class B and C) in studies by All VRC plasma concentrations were determined using a
Wang et al. (Wang et al., 2018a, 2018b). Elevated VRC concen- validated high-performance liquid chromatography–tandem mass
trations may increase the risk of adverse reactions such as spectrometry method (HPLC-MS/MS). D3-voriconazole was used
neurotoxicity, visual disturbances, and potential hepatotoxicity as the internal standard. Chromatographic separation was
(Dolton and McLachlan, 2014). performed on a Hypersil GOLD C18 column (50 mm  4.6 mm,
The non-linear PK, significant inter-individual variability, and 5 mm; Thermo Scientific) with an isocratic mobile phase consisting
narrow therapeutic range of VRC (Dolton et al., 2014) may of A (aqueous phase: 0.1% formic acid) and B (organic phase:
complicate its optimal use in ICU patients and further affect the acetonitrile) (A:B = 60:40, v/v), column temperature of 30  C, flow
prognosis of critically ill patients with liver dysfunction. Therefore, rate of 0.4 ml/min, and a total run time of 5.5 min. The multiple
it is vital to refine VRC therapy to improve clinical outcomes for reaction monitoring (MRM) transitions with an electrospray
these patients. However, it appears that data on the PK and dosage ionization source in the positive mode, m/z 350.0 → 281.0 for
optimization of intravenous VRC in critically ill patients with liver VRC and 353.1 → 284.1 for D3-voriconazole, were conducted on a
dysfunction are limited. The main aim of this prospective study triple-quadrupole tandem mass spectrometer to detect VRC. The
was to characterize the PK of intravenous VRC in ICU patients with plasma sample was pretreated by precipitating with acetonitrile.
liver dysfunction by non-compartmental analysis. The linearity range was 0.10 to 30.00 mg/ml. The intra-day and
inter-day precisions were within 1.23–1.95% and 1.73–2.57%,
Patients and methods respectively. The mean extraction recovery ranged from 101.08%
to 104.92%. A matrix effect on VRC was not observed with this
Study population method. Stability tests showed that VRC was stable in plasma at
room temperature for 24 h and at 20  C in three repeated freeze–
A prospective observational clinical study was conducted from thaw cycles, with tested recoveries between 96.30% and 102.06%.
November 2018 to August 2019 in the Department of Critical Care
Medicine, the First Affiliated Hospital of Sun Yat-sen University. DNA purification and CYP2C19 genotyping
The study was approved by the Ethics Committee of the First
Affiliated Hospital of Sun Yat-sen University (approval number A blood sample (1 ml) was drawn from each enrolled non-
201848). All patients with liver dysfunction aged 18 years or older liver transplant patient for genotype detection. DNA was
receiving intravenous VRC were eligible to be enrolled in the study, purified with the Nucleic Acid Extraction Reagent (Shanghai
and signed informed consent was obtained from participants BaiO Technology Co., Ltd, Shanghai, China). The genotyping of
themselves or their legal guardians before enrollment. The Child– deficient alleles CYP2C19*2 and *3 was performed using the DNA
Pugh classification system was used to categorize the liver function microarray chip method with CYP2C19 Gene Detection Kit
of the patients into mild (Child–Pugh score of 5–6), moderate (Shanghai BaiO Technology Co., Ltd, Shanghai, China). CYP2C19
(Child–Pugh score of 7–9), or severe (Child–Pugh score >9) degree genotypes were classified into six categories: *1/*1, *1/*2, *1/*3,
of hepatic impairment. The exclusion criteria were as follows: (1) *2/*2, *2/*3, and *3/*3.
concomitant use of strong CYP450 enzyme inhibitors or inducers
(e.g., rifampicin and phenytoin); (2) application of extracorporeal Pharmacokinetic analysis
membrane oxygenation; (3) absence of plasma VRC levels; (4)
absence of important dosing information or clinical data (missing A non-compartmental analysis was performed using Phoenix
data). WinNonlin software (version 7.0; Certara LP, Pharsight, St. Louis,
MO, USA) to calculate the PK parameters of PK curve 1 and PK curve
Study design 2. The major parameters included the terminal half-life (t1/2), peak
concentration (Cmax), time to reach Cmax (Tmax), apparent steady-
The initial intravenous (approximately 1-h infusion) VRC state volume of distribution (Vss), clearance (CL), and the area
(VFEND, Pfizer) dosing regimen comprised a loading dose of 300 under the concentration–time curve from 0 h to 12 h (AUC0–12).
X.- Lin et al. / International Journal of Infectious Diseases 93 (2020) 345–352 347

Statistical analysis PK curve 2 was drawn at a median of the 15th dose,12th dose, and
11th dose for Child–Pugh class A (n = 3), B (n = 5), and C (n = 4)
Continuous variables were presented as the median and patients, respectively. As the degree of hepatic impairment
interquartile range (IQR) and categorical variables were reported increased, the CL of VRC of PK curve 1 and PK curve 2 decreased
as frequencies and percentages. The correlations between AUC0–12 (Child–Pugh class A > B > C) and the t1/2 and the AUC0–12 of PK curve
of PK curve 2 and the corresponding C0 and Cmax were evaluated 2 increased (Child–Pugh class A < B < C) (Table 3 and Figure 3).
using a simple linear regression model. One-way analysis of However, the differences in these PK parameters were not
variance (ANOVA) with Bonferroni post hoc test was used to statistically significant among the three Child–Pugh classes, except
compare the difference in first C0 among the Child–Pugh classes. for CL in PK curve 1 (p = 0.012). Additionally, the CL (median) in PK
Simple linear regression analysis was used to evaluate the curve 2 was lower than that in PK curve 1 for patients of all three
correlations between PK parameters and the clinical character- Child–Pugh classes (Table 3 and Figure 3).
istics and biochemical indices of the patients. A two-sided p-value The median (IQR) first C0 of the 17 patients was 5.27 (2.61) mg/
of <0.05 was considered statistically significant. The statistical ml. The median sampling time for the first C0 was day 5 (range day
analysis was performed using IBM SPSS software version 24.0 (IBM 4–7). Only two C0 values were less than the lower limit of the
Corp., Armonk, NY, USA). therapeutic window (2–6 mg/ml), whereas 29.4% (5/17) of the first
C0 exceeded the upper limit of 6 mg/ml. Furthermore, the first C0
Results was elevated with increased degree of hepatic impairment (Child–
Pugh class A < B < C), but only the difference in C0 between Child–
Patient characteristics Pugh class C and A patients was significant (p = 0.030; Figure 4).
The median (IQR) of the first C0 in Child–Pugh class A (n = 4), B
A total of 17 patients were included in the study. PK curve 1 was (n = 8), and C (n = 5) patients was 2.41 (3.57), 5.22 (2.06), and 5.96
drawn within one dosing interval of the first dose for all 17 (4.18) mg/ml, respectively; the median sampling time for the first
patients; PK curve 2 was drawn within one dosing interval after a C0 was day 5, day 5, and day 4, respectively.
minimum of seven doses (range 7 to 15 doses; median 12 doses) for The significant correlations between the PK parameters and the
12 patients (PK profiles are shown in Figure 1; and the actual clinical characteristics and biochemical indices of the patients,
sampling times and number of samples per subject are presented determined by simple linear regression, are summarized in Table 4
in Supplementary MaterialTable S1). The demographic and and Figure 5. The t1/2 of PK curve 2 extended with elevated
clinical characteristics of the 17 patients (PK curve 1) and the 12 international normalized ratio (INR) (r2 = 0.510); AUC0–12 of PK
patients (PK curve 2) are summarized in Table 1. The range of actual curve 2 increased with elevated aspartate aminotransferase (AST)
body weight of the 17 patients was 48–87 kg (median 58 kg). The (r2 = 0.612) and total bilirubin (TBIL) (r2 = 0.515). Vss of PK curve 1
subjects were divided into three categories based on the degree of was positively correlated with actual body weight (r2 = 0.450), and
liver dysfunction: four were Child–Pugh class A, eight were class B, the correlation was significantly improved (r2 = 0.707) when an
and five were class C. Four categories of CYP2C19 genotypes were outlier of Vss (309.21 l) from patient 3 was omitted (Figure 5). In
observed in the 12 non-liver transplant patients: *1/*1 (n = 5), *1/*2 addition, there was a negative correlation between AUC0–12 of PK
(n = 5), *2/*2 (n = 1), and *2/*3 (n = 1). The frequencies of deficient curve 1 and body weight (r2 = 0.614). The study did not observe
alleles CYP2C19*2 and*3 were 33.3% and 4.2%, respectively. significant associations between the PK parameters and other
clinical characteristics (e.g., APACHE II score, SOFA score at
Pharmacokinetics results baseline, CYP2C19 genotype, and concomitant use of PPIs
(esomeprazole vs. pantoprazole) and VRC).
The PK parameters of intravenous VRC in the 17 patients (PK curve
1) and the 12 patients (PK curve 2), as well as in the different Child– Discussion
Pugh classes, are presented in Tables 2 and 3. The Tmax, Cmax, Vss, and
AUC0–12 of PK curve 1 did not include patient 14, because Cmax was To date, the PK profile of intravenous VRC in critically ill patients
not obtained for this patient. As shown in Figure 2, there were good with liver dysfunction has not been studied. The current
correlations between AUC0–12 of PK curve 2 and the corresponding prospective study of 17 ICU patients with hepatic impairment
C0 and Cmax (r2 = 0.951 and 0.963, respectively; both p < 0.001). receiving a standard VRC dosing regimen, investigated the PK

Figure 1. Plasma concentration vs. time curves of intravenous voriconazole in patients with liver dysfunction. (A) Individual plasma concentration vs. time curves (PK curve 1)
for 17 patients during an intravenous infusion (300 mg, approximately 1-h infusion) dosing interval of the first dose. (B) Individual plasma concentration vs. time curves (PK
curve 2) for 12 patients during an intravenous infusion (200 mg, approximately 1-h infusion) dosing interval of a minimum of seven doses (range 7 to 15 doses; median 12
doses). Abbreviation: PK, pharmacokinetics.
348 X.- Lin et al. / International Journal of Infectious Diseases 93 (2020) 345–352

Table 1
Demographic and clinical data; median (IQR, range).

Characteristic PK curve 1 (n = 17) PK curve 2 (n = 12)


First C0 (mg/ml) 5.27 (2.61, 1.72–10.07) 5.42 (4.37, 1.87–10.07)
Sampling time for first C0 (days) 5 (1, 4–7) 5 (1, 4–5)
SOFA score at baseline 10 (7, 5–21) 11 (7, 6–21)
APACHE II score 23 (13, 8–29) 24 (11, 9–29)
Child–Pugh classification, n (%)
A; B; C 4 (23.5); 8 (47.1); 5 (29.4) 3 (25.0); 5 (41.7); 4 (33.3)
Demographic data
Sex, male/female, n/n 13/4 9/3
Age (years) 48 (31, 33–89) 51 (28, 33–72)
Actual body weight (kg) 58 (17, 48–87) 55 (14, 48–87)
Etiology, n (%)
Hepatocellular carcinoma 1 (5.9) 1 (8.3)
Chronic hepatitis B 2 (11.8) 2 (16.7)
Hepatitis B cirrhosis 2 (11.8) 1 (8.3)
Cholelithiasis 2 (11.8) 1 (8.3)
Transplanted 5 (29.4) 3 (25.0)
Sepsis-induced 3 (17.6) 2 (16.7)
Others 2 (11.8) 2 (16.7)
CYP2C19 genotype, n (%)a
*1/*1 5 (41.7) 3 (33.3)
*1/*2 5 (41.7) 5 (55.6)
*2/*2 1 (8.3) 0 (0.0)
*2/*3 1 (8.3) 1 (11.1)
Concomitant medication, n (%)
Esomeprazole 9 (52.9) 6 (50.0)
Pantoprazole 8 (47.1) 6 (50.0)
Biochemical indicesb
INR 1.39 (0.61, 1.05–2.36) 1.36 (0.36, 0.94–2.51)
ALT (U/l) 29 (63, 1–557) 18 (31, 2–250)
AST (U/l) 76 (90, 27–1322) 49 (67, 14–145)
TBIL (mmol/l) 132.6 (217.1, 9.9–439.5) 93.0 (253.3, 9.3–349.3)
ALB (g/l) 36.1 (11.0, 24.4–52.7) 34.6 (8.2, 28.8–45.5)
PLT (109/l) 62 (105, 14–424) 58 (133, 10–503)
SCr (mmol/l) 96 (96, 47–356) 75 (49, 57–305)

ALB, albumin; ALT, alanine aminotransferase; APACHE II score, Acute Physiology and Chronic Health Evaluation II score; AST, aspartate aminotransferase; C0, trough
concentration; INR, international normalized ratio; IQR, interquartile range; PK, pharmacokinetics; PLT, platelets; SCr, serum creatinine; SOFA score, sequential organ failure
assessment score; TBIL, total bilirubin.
a
Data are available for 12 patients because CYP2C19 genotyping was not performed for the other five patients with a history of liver transplantation.
b
Biochemical indices were measured on the day of sampling.

characteristics of intravenous VRC in different Child–Pugh classes Table 2


using non-compartmental analysis. PK parameters of intravenous voriconazole of PK curve 1 and PK curve 2; median
In the present study, a simple and reliable HPLC-MS/MS assay (IQR, range).
was developed to determine VRC plasma concentrations. Accord- PK parameters PK curve 1 (n = 17)a PK curve 2 (n = 12)
ing to the United States Food and Drug Administration bioanalyt-
t1/2 (h) 14.4 (12.4, 8.3–149.7) 31.3 (26.1, 5.9–180.0)
ical method validation guidance (U.S. Food and Drug Tmax (h) 1.01 (0.27, 0.75–1.20) 1.08 (0.16, 0.77–1.18)
Administration, 2001), this method is fully validated for specificity, Cmax (mg/ml) 4.90 (3.06, 1.92–9.12) 9.63 (3.11, 4.83–16.86)
linearity, precision, accuracy, recovery, matrix effect, and stability. Vss (l) 119.85 (91.29, 55.45– 112.57 (141.47, 52.35–
309.21) 554.97)
In some previously developed LC-MS/MS methods (Al-Ghobashy
CL (l/h) 5.09 (2.78, 0.60–9.57) 2.33 (1.13, 1.18–6.75)
et al., 2018; Prommas et al., 2017), fluconazole was used as the AUC0–12 (mgh/ 26.55 (14.21, 13.20–49.06) 86.29 (34.88, 29.61–169.85)
internal standard. Considering that VRC might be used after ml)
fluconazole in clinical settings, we used deuterated VRC
AUC0–12, area under the concentration–time curve from 0 h to 12 h; CL, clearance;
(D3-voriconazole) as the internal standard, which eliminated Cmax, peak concentration; IQR, interquartile range; PK, pharmacokinetics; t1/2,
the interference of drugs including fluconazole on the detection of terminal half-life; Tmax, time to reach Cmax; Vss, apparent steady-state volume of
the internal standard. The adopted method was validated to be distribution.
a
Data on Tmax, Cmax, Vss, and AUC0–12 of PK curve 1 were available for 16 patients;
accurate and precise, with high extraction recovery and no matrix
data for patient 14 were not included, because Cmax was not obtained for this
effect, and can be applied to clinical TDM for VRC. patient.
The estimated median CL (2.33 l/h) of PK curve 2 in the present
study is lower than the values reported in several studies that were corresponding Cmax (r2 = 0.963), indicating that both C0 and Cmax
not focused on patients with liver dysfunction (4.28–6.95 l/h) are good surrogates for measuring VRC plasma exposure (AUC). In
(Chen et al., 2015; Pascual et al., 2012; Wang et al., 2014a). the current study, the first C0 determined on day 5 (median) was as
Moreover, the t1/2 of PK curve 2 was 31.3 h (median), which is high as 5.27 (2.61) mg/ml; only two C0 values were less than the
substantially longer than that of healthy volunteers (6 h) reported lower target of the therapeutic window (2–6 mg/ml), but 29.4%
in the early PK studies (Jeu et al., 2003). Consistent with previous (5/17) exceeded the upper limit of 6 mg/ml. Similarly, under a
findings (Chen et al., 2015; Han et al., 2010; Johnson et al., 2010; standard VRC maintenance dose of 200 mg twice daily, a high C0
Wang et al., 2015), there was a close correlation between AUC0–12 was also observed in patients with liver dysfunction by Gao et al.
and the corresponding C0 (r2 = 0.951) in the present study. We also (Gao et al., 2018) and Wang et al. (Wang et al., 2018b). These results
found that AUC0–12 was significantly correlated with the indicate that decreased liver function could substantially reduce
X.- Lin et al. / International Journal of Infectious Diseases 93 (2020) 345–352 349

Table 3
PK parameters of intravenous voriconazole of PK curve 1 and PK curve 2 in the different Child–Pugh classes; median (IQR).

PK parameters PK curve 1 (n = 17), Child–Pugh class PK curve 2 (n = 12), Child–Pugh class


a
A (n = 4) B (n = 8) C (n = 5) A (n = 3) B (n = 5) C (n = 4)
t1/2 (h) 12.6 (5.3) 20.7 (12.3) 14.5 (85.5) 24.4 (NA) 29.1 (20.4) 60.7 (129.4)
Tmax (h) 1.00 (NA) 1.02 (0.21) 0.78 (0.25) 1.08 (NA) 1.00 (0.28) 1.15 (0.13)
Cmax (mg/ml) 5.79 (NA) 4.36 (2.18) 6.64 (5.41) 7.76 (NA) 9.13 (3.98) 10.22 (5.64)
Vss (l) 85.58 (NA) 129.40 (83.16) 99.65 (150.89) 83.42 (NA) 124.89 (55.60) 159.16 (414.65)
CL (l/h) 8.60 (4.13) 5.21 (2.13) 4.72 (3.37) 3.31 (NA) 2.54 (1.06) 2.04 (0.77)
AUC0–12 (mgh/ml) 24.91 (NA) 23.37 (12.99) 28.60 (20.25) 60.45 (NA) 78.83 (35.62) 98.04 (59.48)

AUC0–12, area under the concentration–time curve from 0 h to 12 h; CL, clearance; Cmax, peak concentration; IQR, interquartile range; NA, not available; PK, pharmacokinetics;
t1/2, terminal half-life; Tmax, time to reach Cmax; Vss, apparent steady-state volume of distribution.
a
Data on Tmax, Cmax, Vss, and AUC0–12 were available for three patients; data for patient 14 were not included, because Cmax was not obtained for this patient.

Figure 2. Correlations between AUC0–12 of PK curve 2 and the corresponding C0 and Cmax (n = 12). (A) AUC0–12 vs. C0. (B) AUC0–12 vs. Cmax. Abbreviations: AUC0–12, area under
the concentration–time curve from 0 h to 12 h; C0, trough concentration; Cmax, peak concentration; PK, pharmacokinetics.

Figure 3. Comparisons for the PK parameters (median) of PK curve 1 and PK curve 2 among Child-Pugh classes.(A) t1/2. (B) CL. (C) AUC0–12. Abbreviations: AUC0–12, area under
the concentration–time curve from 0 h to 12 h; C-P, Child-Pugh; CL, clearance; PK, pharmacokinetics; t1/2, terminal half-life.

VRC metabolism, resulting in a significant decrease in CL and a Considering that elevated VRC exposure may increase the risk of
significant prolongation of t1/2, ultimately leading to high plasma developing adverse reactions, dosing regimens should be adjusted
exposure. based on Child–Pugh classes for those ICU patients with liver
Although not all of the differences in PK parameters among the dysfunction.
three Child–Pugh classes were statistically significant due to the Saturable metabolism leads to non-linear PK of VRC (Theur-
limited sample size of this study, as the degree of hepatic etzbacher et al., 2006). However, non-linear PK was not observed
impairment increased (Child–Pugh class A < B < C), CL of VRC in some studies, probably owing to the lack of intensive sampling
decreased and t1/2 extended; as a result, AUC0–12 and the first C0 (Pascual et al., 2012; Wang et al., 2014a). In the present study, the
elevated. The CL (PK curve 2) of Child–Pugh class B and C patients CL (median) of PK curve 2 was lower than that of PK curve 1 in all
in our study were decreased to 2.54 and 2.04 l/h, respectively. three Child–Pugh classes, which could be attributed to the fact that
Recently, lower CL of 0.99 l/h in Child–Pugh class C cirrhotic the largely decreased metabolism of VRC in patients with liver
patients was reported in a retrospective study by Wang et al. dysfunction make it more susceptible to saturable metabolism. The
(Wang et al., 2019). In addition, t1/2 of Child–Pugh class C patients non-linear PK, one of the important sources for the wide individual
in their study was extended to 111.7 h, which is nearly twice as long variability of VRC, would further complicate the optimal use of VRC
as the value of 60.7 h (PK curve 2) estimated in our study. in patients with liver disease.
350 X.- Lin et al. / International Journal of Infectious Diseases 93 (2020) 345–352

It should be mentioned that CYP2C19 genotyping was


performed only for the 12 patients without a history of liver
transplantation. If the CYP2C19 genotypes differ between a donor
and a recipient, the expression level of CYP2C19 in liver graft tissue
depends on the donor (Chiu et al., 2015), indicating that the
CYP2C19 genotypes detected from peripheral blood of recipients
cannot replace the genotypes and phenotypes of CYP2C19 in the
liver graft. Therefore, CYP2C19 genotyping was not performed for
the patients with a history of liver transplantation (patients 4, 6, 7,
10, and 11) in our study. The distribution of CYP2C19 alleles varies
in different race/ethnic populations, including in the Asian
population (Zhou et al., 2019). The frequencies of CYP2C19*2, *3,
and *17 in East Asians are 28.4%, 7.3%, and 2.1%, respectively, but
those in Central/South Asians are 27.0%, 1.6%, and 17.1%,
respectively (PharmGKB). In addition, Chuwongwattana et al.
(Chuwongwattana et al., 2016) reported that the frequencies of
CYP2C19*2 and *3 in Thai patients were 25.7% and 4.8%,
respectively, but CYP2C19*17 was not found in their study. The
frequencies of CYP2C19*2 and *3 (33.3% and 4.2%, respectively) in
the 12 Chinese patients in our study were roughly in line with the
data for Chinese patients reported in several previous studies
(29.2–31.2% and 4.3–5.2%, respectively) (Lin et al., 2018; Miao et al.,
Figure 4. Distribution of the first C0 among Child-Pugh classes. The Bonferroni 2019; Zhong et al., 2017; Zhuo et al., 2018). In a previous study, the
correction was made for pairwise comparisons following the one-way ANOVA prevalences of CYP2C19*3 in Chinese liver transplant recipients and
(p = 0.032). Data are expressed as the median (interquartile range). Abbreviations:
donors were reported to be as high as 24.2% and 26.9%,
C0, trough concentration; C-P, Child-Pugh.
respectively, while no CYP2C19*2 was observed in either recipients
or donors (Zhu et al., 2016). This large difference in the distribution
As the actual body weight increased, Vss of PK curve 1 increased of CYP2C19 alleles needs to be further studied.
(r2 = 0.707, n = 15) and AUC0–12 of PK curve 1 decreased (r2 = 0.614, In recent years, CYP2C19 polymorphisms have been identified to
n = 16). This supports the intravenous administration of a loading be an important determinant of the wide PK variability of VRC (Lee
dose of VRC for adult ICU patients with liver dysfunction based on et al., 2012; Li et al., 2017; Lin et al., 2018; Wang et al., 2014a). The
their actual body weight. However, an actual body weight-adjusted loss-of-function alleles CYP2C19*2 and *3 have been associated
dosing regimen may not be feasible for obese patients, as this with increased C0 of VRC (Chuwongwattana et al., 2016;
would lead to high VRC concentrations (Davies-Vorbrodt et al., Lamoureux et al., 2016). Several studies have found the CL of
2013; Koselke et al., 2012). A dosing regimen based on adjusted VRC in poor metabolizers (CYP2C19*2/*2, *2/*3, *3/*3) to be low (Lin
body weight has been proposed (Davies-Vorbrodt et al., 2013; et al., 2018; Wang et al., 2014a), resulting in significantly higher
Koselke et al., 2012; Richards et al., 2017), and further studies are plasma C0 compared with those in both intermediate metabolizers
needed. In the current study, no obese patient (body mass (CYP2C19*1/*2, *1/*3) (Lin et al., 2018; Wang et al., 2014b) and
index 30 kg/m2) was observed. The influence of obesity on the normal metabolizers (CYP2C19*1/*1) (Chuwongwattana et al.,
PK of VRC in patients with liver dysfunction should be investigated 2016; Lin et al., 2018; Wang et al., 2014b). However, the effect of
in the future. Additionally, t1/2 (PK curve 2) extended with elevated CYP2C19 polymorphisms on the PK parameters of VRC was not
INR (r2 = 0.510), and AUC0–12 (PK curve 2) increased with elevated observed in patients with liver dysfunction by Tang et al. in their
AST (r2 = 0.612) and TBIL (r2 = 0.515). These findings also reflect the recent retrospective study (Tang et al., 2019), and they attributed
effect of liver function on the PK of VRC. Nevertheless, the the discrepancy to the decrease in activity of CYP450 enzymes. The
correlations between other PK parameters and indices of liver present study also did not find any significant associations
function (including INR, AST, and TBIL) were significant but weak in between the PK parameters and CYP2C19 genotypes (carrying at
our study. Further research is required to identify the effects of least one deficient allele (*2 or *3) or not), but it should be
liver function indices on PK parameters of VRC for patients with mentioned that a subgroup analysis in the different Child–Pugh
liver disease. classes could not be performed to evaluate the effect of CYP2C19

Table 4
Correlations between PK parameters and the clinical characteristics and biochemical indices of the patients.

Variablesa First C0 (n = 17) PK curve 1 (n = 17) PK curve 2 (n = 12)

t1/2b CL AUC0–12c Vssc t1/2d CL AUC0–12 Vssd


Body weight 0.243 (+) 0.614 ( ) 0.450 (+)
INR 0.234 (+) 0.303 (+) 0.510 (+)
AST 0.355 (+) 0.361 ( ) 0.612 (+)
ALB 0.312 ( )
TBIL 0.312 (+) 0.307 ( ) 0.515 (+)

ALB, albumin; AST, aspartate aminotransferase; AUC0–12, area under the concentration–time curve from 0 h to 12 h; C0, trough concentration; CL, clearance; INR, international
normalized ratio; PK, pharmacokinetics; t1/2, terminal half-life; TBIL, total bilirubin; Vss, apparent steady-state volume of distribution.
a
Statistically significant r2 for simple linear regression between the two variables is shown, and symbols in parentheses indicate a positive (+) or negative ( ) association.
b
Analyses were available for 16 patients; data for patient 5 were excluded, because the t1/2 (149.7 h) of this patient was identified as an extreme outlier.
c
Analyses were available for 16 patients; data for patient 14 were not included, because Vss and AUC0–12 were not available for this patient.
d
Analyses were available for 11 patients; data for patient 3 were excluded, because both t1/2 (180.0 h) and Vss (554.97 l) of this patient were identified as extreme outliers.
X.- Lin et al. / International Journal of Infectious Diseases 93 (2020) 345–352 351

Author contributions

Study conception and design: Xiao Chen, Chang-jie Cai. Data


acquisition: Fa Huang, Li Tong, Yan-zhe Xia, Xiao-guang Hu. Data
analysis and interpretation: Xiao-bin Lin, Guo-ping Zhong.
Determination of plasma concentrations: Jing-jing Wu. DNA
extraction and detection: Jia Li, Tao Liang. Draft preparation:
Xiao-bin Lin, Fa Huang. Draft revision: Xiao-man Liu. Critical
review: All authors.

Funding

This work was supported by the National Key R&D Program of


China (No. 2017YFC0909904) and the Guangzhou Science and
Technology Program (No. 201704020153).

Ethical approval

This study was approved by the Ethics Committee of the First


Affiliated Hospital of Sun Yat-sen University (approval number 201848).

Conflict of interest

Figure 5. Correlation between Vss of PK curve 1 and actual body weight. Main The authors declare that they have no conflict of interest.
figure, r2 = 45.0% when Vss and body weight were correlated for 16 patients. Inset,
r2 = 70.7% when Vss and body weightwere correlated for 15 patients, with an outlier Acknowledgements
of Vss (309.21 l) from patient 3 omitted. Abbreviations: PK, pharmacokinetics; Vss,
apparent steady-state volume of distribution.
We appreciate Jia-zhi Chen and Rui Dai for their assistance in
the collection of clinical data and blood samples.
genotypes on PK parameters owing to the limited sample size, so
further studies are warranted. Appendix A. Supplementary data
All of the patients in the present study were co-administered
CYP2C19 inhibitor PPIs (esomeprazole or pantoprazole) during Supplementary material related to this article can be found, in
VRC therapy. The effects of PPIs on VRC PK depend on the type of the online version, at doi:https://doi.org/10.1016/j.ijid.2020.02.041.
PPI used (Qi et al., 2017; Yan et al., 2018; Yasu et al., 2016). Co-
administration with esomeprazole significantly increased VRC References
plasma C0, whereas there was no significant correlation between
Alffenaar JW, de Vos T, Uges DR, Daenen SM. High voriconazole trough levels in
plasma C0 and the use of pantoprazole (Yan et al., 2018). In the
relation to hepatic function: how to adjust the dosage?. Br J Clin Pharmacol
current study, it was difficult to compare the differences in PK 2009;67(2):262–3.
parameters between the co-administration of esomeprazole and Al-Ghobashy MA, Kamal SM, El-Sayed GM, Attia AK, Nagy M, ElZeiny A, et al.
Determination of voriconazole and co-administered drugs in plasma of
pantoprazole, since it was not possible to perform a subgroup
pediatric cancer patients using UPLC-MS/MS: a key step towards personalized
analysis due to the limited sample size. As the concomitant use of therapeutics. J Chromatogr B Analyt Technol Biomed Life Sci 2018;1092:489–98.
the two drugs in the ICU is fairly common, the influence of PPIs on Chen J, Yang Q, Huang J, Li L. Risk factors for invasive pulmonary aspergillosis and
VRC PK should be explored in patients with liver dysfunction. hospital mortality in acute-on-chronic liver failure patients: a retrospective-
cohort study. Int J Med Sci 2013;10(12):1625–31.
The main limitation of this study is the small sample size, which Chen W, Xie H, Liang F, Meng D, Rui J, Yin X, et al. Population pharmacokinetics in
limited the power to evaluate the differences in PK parameters china: the dynamics of intravenous voriconazole in critically ill patients with
among the different Child–Pugh classes. Secondly, data on CYP2C19 pulmonary disease. Biol Pharm Bull 2015;38(7):996–1004.
Chiu KW, Nakano T, Chen KD, Hsu LW, Lai CY, Huang CY, et al. Cytochrome P450 in
genotypes were only available for 12 patients, thus a subgroup living donor liver transplantation. J Biomed Sci 2015;22:32.
analysis could not be performed in our study. Therefore, further Chuwongwattana S, Jantararoungtong T, Chitasombat MN, Puangpetch A, Prommas
investigations of the effects of CYP2C19 polymorphisms on the PK S, Dilokpattanamongkol P, et al. A prospective observational study of CYP2C19
polymorphisms and voriconazole plasma level in adult Thai patients with
of VRC in patients with hepatic impairment are required. invasive aspergillosis. Drug Metab Pharmacokinet 2016;31(2):117–22.
Additionally, dosing adjustment strategies could not be provided Davies-Vorbrodt S, Ito JI, Tegtmeier BR, Dadwal SS, Kriengkauykiat J. Voriconazole
in the current study due to the lack of data on plasma exposure serum concentrations in obese and overweight immunocompromised patients:
a retrospective review. Pharmacotherapy 2013;33(1):22–30.
(AUC0–12 of PK curve 2) of patients with normal liver function for
Dolton MJ, McLachlan AJ. Voriconazole pharmacokinetics and exposure-response
comparison. Recommending adequate dosing regimens for ICU relationships: assessing the links between exposure, efficacy and toxicity. Int J
patients with liver dysfunction is challenging and further studies Antimicrob Agents 2014;44(3):183–93.
are needed. Despite the limitations, the present study may provide Dolton MJ, Mikus G, Weiss J, Ray JE, McLachlan AJ. Understanding variability with
voriconazole using a population pharmacokinetic approach: implications for
a theoretical basis for the clinical use of VRC in ICU patients with optimal dosing. J Antimicrob Chemother 2014;69(6):1633–41.
liver dysfunction and serve as a reference for further research. Gao J, Zhang Q, Wu Y, Li Y, Qi T, Zhu C, et al. Improving survival of acute-on-chronic
In conclusion, the CL of VRC in patients with liver dysfunction liver failure patients complicated with invasive pulmonary aspergillosis. Sci Rep
2018;8(1):876.
(especially Child–Pugh class C) largely decreased and t1/2 extended, Han K, Capitano B, Bies R, Potoski BA, Husain S, Gilbert S, et al. Bioavailability and
resulting in high plasma exposure. For these ICU patients, dosing population pharmacokinetics of voriconazole in lung transplant recipients.
regimens of intravenous VRC should be adjusted based on Child– Antimicrob Agents Chemother 2010;54(10):4424–31.
Hyland R, Jones BC, Smith DA. Identification of the cytochrome P450 enzymes
Pugh classes, and plasma concentrations should be closely involved in the N-oxidation of voriconazole. Drug Metab Dispos 2003;31
monitored to avoid serious adverse events. (5):540–7.
352 X.- Lin et al. / International Journal of Infectious Diseases 93 (2020) 345–352

Jeu L, Piacenti FJ, Lyakhovetskiy AG, Fung HB. Voriconazole. Clin Ther 2003;25 population pharmacokinetic approach. Basic Clin Pharmacol Toxicol 2019;125
(5):1321–81. (1):34–43.
Jeurissen S, Vogelaers D, Sermijn E, Van Dycke K, Geerts A, Van Vlierberghe H, et al. Theuretzbacher U, Ihle F, Derendorf H. Pharmacokinetic/pharmacodynamic profile
Invasive aspergillosis in patients with cirrhosis, a case report and review of the of voriconazole. Clin Pharmacokinet 2006;45(7):649–63.
last 10 years. Acta Clin Belg 2013;68(5):368–75. U.S. Food and Drug Administration. Guidance for industry bioanalytical method
Johnson HJ, Han K, Capitano B, Blisard D, Husain S, Linden PK, et al. Voriconazole validation. 2001.
pharmacokinetics in liver transplant recipients. Antimicrob Agents Chemother Verbeeck RK. Pharmacokinetics and dosage adjustment in patients with hepatic
2010;54(2):852–9. dysfunction. Eur J Clin Pharmacol 2008;64(12):1147–61.
Koselke E, Kraft S, Smith J, Nagel J. Evaluation of the effect of obesity on voriconazole Veringa A, Ter Avest M, Span LF, van den Heuvel ER, Touw DJ, Zijlstra JG, et al.
serum concentrations. J Antimicrob Chemother 2012;67(12):2957–62. Voriconazole metabolism is influenced by severe inflammation: a prospective
Koulenti D, Garnacho-Montero J, Blot S. Approach to invasive pulmonary study. J Antimicrob Chemother 2017;72(1):261–7.
aspergillosis in critically ill patients. Curr Opin Infect Dis 2014;27(2):174–83. Wang T, Chen S, Sun J, Cai J, Cheng X, Dong H, et al. Identification of factors
Lamoureux F, Duflot T, Woillard JB, Metsu D, Pereira T, Compagnon P, et al. Impact of influencing the pharmacokinetics of voriconazole and the optimization of
CYP2C19 genetic polymorphisms on voriconazole dosing and exposure in adult dosage regimens based on Monte Carlo simulation in patients with invasive
patients with invasive fungal infections. Int J Antimicrob Agents 2016;47 fungal infections. J Antimicrob Chemother 2014a;69(2):463–70.
(2):124–31. Wang T, Xie J, Wang Y, Zheng X, Lei J, Wang X, et al. Pharmacokinetic and
Lee S, Kim BH, Nam WS, Yoon SH, Cho JY, Shin SG, et al. Effect of CYP2C19 pharmacodynamic properties of oral voriconazole in patients with invasive
polymorphism on the pharmacokinetics of voriconazole after single and fungal infections. Pharmacotherapy 2015;35(9):797–804.
multiple doses in healthy volunteers. J Clin Pharmacol 2012;52(2):195–203. Wang T, Yan M, Tang D, Dong Y, Zhu L, Du Q, et al. Pharmacokinetics modeling and
Li ZW, Peng FH, Yan M, Liang W, Liu XL, Wu YQ, et al. Impact of CYP2C19 genotype simulation of voriconazole dosing and safety in patients with liver cirrhosis. ,
and liver function on voriconazole pharmacokinetics in renal transplant doi:http://dx.doi.org/10.1101/563528.
recipients. Ther Drug Monit 2017;39(4):422–8. Wang T, Yan M, Tang D, Xue L, Zhang T, Dong Y, et al. A retrospective, multicenter
Lin XB, Li ZW, Yan M, Zhang BK, Liang W, Wang F, et al. Population pharmacokinetics study of voriconazole trough concentrations and safety in patients with Child-
of voriconazole and CYP2C19 polymorphisms for optimizing dosing regimens in Pugh class C cirrhosis. J Clin Pharm Ther 2018a;43(6):849–54.
renal transplant recipients. Br J Clin Pharmacol 2018;84(7):1587–97. Wang T, Yan M, Tang D, Xue L, Zhang T, Dong Y, et al. Therapeutic drug monitoring
Miao Q, Tang JT, van Gelder T, Li YM, Bai YJ, Zou YG, et al. Correlation of CYP2C19 and safety of voriconazole therapy in patients with Child-Pugh class B and C
genotype with plasma voriconazole exposure in South-western Chinese Han cirrhosis: a multicenter study. Int J Infect Dis 2018b;72:49–54.
patients with invasive fungal infections. Medicine (Baltimore) 2019;98(3) Wang T, Zhu H, Sun J, Cheng X, Xie J, Dong H, et al. Efficacy and safety of voriconazole
e14137. and CYP2C19 polymorphism for optimised dosage regimens in patients with
Pappas PG, Kauffman CA, Andes DR, Clancy CJ, Marr KA, Ostrosky-Zeichner L, et al. invasive fungal infections. Int J Antimicrob Agents 2014b;44(5):436–42.
Clinical practice guideline for the management of Candidiasis: 2016 update by Yamada T, Imai S, Koshizuka Y, Tazawa Y, Kagami K, Tomiyama N, et al. Necessity for
the infectious diseases society of America. Clin Infect Dis 2016;62(4):e1–50. a significant maintenance dosage reduction of voriconazole in patients with
Pascual A, Csajka C, Buclin T, Bolay S, Bille J, Calandra T, et al. Challenging severe liver cirrhosis (Child-Pugh class C). Biol Pharm Bull 2018;41(7):1112–8.
recommended oral and intravenous voriconazole doses for improved efficacy Yan M, Wu ZF, Tang D, Wang F, Xiao YW, Xu P, et al. The impact of proton pump
and safety: population pharmacokinetics-based analysis of adult patients with inhibitors on the pharmacokinetics of voriconazole in vitro and in vivo. Biomed
invasive fungal infections. Clin Infect Dis 2012;55(3):381–90. Pharmacother 2018;108:60–4.
Patterson TF, Thompson 3rd GR, Denning DW, Fishman JA, Hadley S, Herbrecht R, Yasu T, Konuma T, Kato S, Kurokawa Y, Takahashi S, Tojo A. Different effects of
et al. Practice guidelines for the diagnosis and management of aspergillosis: lansoprazole and rabeprazole on the plasma voriconazole trough levels in
2016 update by the infectious diseases society of America. Clin Infect Dis allogeneic hematopoietic cell transplant recipients. Ann Hematol 2016;95
2016;63(4):e1–e60. (11):1845–51.
PharmGKB. Gene-specific information tables for CYP2C19: CYP2C19 frequency Zhong Z, Hou J, Li B, Zhang Q, Liu S, Li C, et al. Analysis of CYP2C19 genetic
table. Available from: https://www.pharmgkb.org/page/cyp2c19RefMaterials. polymorphism in a large ethnic hakka population in Southern China. Med Sci
[Accessed 16 January 2020]. Monit 2017;23:6186–92.
Prommas S, Puangpetch A, Jenjirattithigarn N, Chuwongwattana S, Jantararoung- Zhou L, Sharma P, Yeo KR, Higashimori M, Xu H, Al-Huniti N, et al. Assessing
tong T, Koomdee N, et al. Development and validation of voriconazole pharmacokinetic differences in Caucasian and East Asian (Japanese, Chinese
concentration by LC-MS-MS: applied in clinical implementation. J Clin Lab and Korean) populations driven by CYP2C19 polymorphism using physiologi-
Anal 2017;31(1). cally-based pharmacokinetic modelling. Eur J Pharm Sci 2019;139:105061.
Qi F, Zhu L, Li N, Ge T, Xu G, Liao S. Influence of different proton pump inhibitors on the Zhu L, Liao S, Wang N, Ge T, Yang J, Xu G, et al. Dose regimens for Chinese adult liver
pharmacokinetics of voriconazole. Int J Antimicrob Agents 2017;49(4):403–9. transplant recipients according to the genetic polymorphisms of CYP2C9,
Richards PG, Dang KM, Kauffman CA, Stalker KL, Sudekum D, Kerr L, et al. CYP2C19, and CYP3A5 in recipients and donors. Int J Clin Pharmacol Ther
Therapeutic drug monitoring and use of an adjusted body weight strategy for 2016;54(8):587–96.
high-dose voriconazole therapy. J Antimicrob Chemother 2017;72(4):1178–83. Zhuo ZL, Xian HP, Long Y, Liu C, Sun YY, Ma YT, et al. Association between CYP2C19
Tang D, Song BL, Yan M, Zou JJ, Zhang M, Zhou HY, et al. Identifying factors affecting and ABCB1 polymorphisms and clopidogrel resistance in clopidogrel-treated
the pharmacokinetics of voriconazole in patients with liver dysfunction: a Chinese patients. Anatol J Cardiol 2018;19(2):123–9.

You might also like