Next Article in Journal
Development of Biotinylated Liposomes Encapsulating Metformin for Therapeutic Targeting of Inflammation-Based Diseases
Previous Article in Journal
Rutin/Sulfobutylether-β-Cyclodextrin as a Promising Therapeutic Formulation for Ocular Infection
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Simultaneously Predicting the Pharmacokinetics of CES1-Metabolized Drugs and Their Metabolites Using Physiologically Based Pharmacokinetic Model in Cirrhosis Subjects

Center of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China
*
Authors to whom correspondence should be addressed.
Pharmaceutics 2024, 16(2), 234; https://doi.org/10.3390/pharmaceutics16020234
Submission received: 13 December 2023 / Revised: 29 January 2024 / Accepted: 2 February 2024 / Published: 5 February 2024

Abstract

:
Hepatic carboxylesterase 1 (CES1) metabolizes numerous prodrugs into active ingredients or direct-acting drugs into inactive metabolites. We aimed to develop a semi-physiologically based pharmacokinetic (semi-PBPK) model to simultaneously predict the pharmacokinetics of CES1 substrates and their active metabolites in liver cirrhosis (LC) patients. Six prodrugs (enalapril, benazepril, cilazapril, temocapril, perindopril and oseltamivir) and three direct-acting drugs (flumazenil, pethidine and remimazolam) were selected. Parameters such as organ blood flows, plasma-binding protein concentrations, functional liver volume, hepatic enzymatic activity, glomerular filtration rate (GFR) and gastrointestinal transit rate were integrated into the simulation. The pharmacokinetic profiles of these drugs and their active metabolites were simulated for 1000 virtual individuals. The developed semi-PBPK model, after validation in healthy individuals, was extrapolated to LC patients. Most of the observations fell within the 5th and 95th percentiles of simulations from 1000 virtual patients. The estimated AUC and Cmax were within 0.5–2-fold of the observed values. The sensitivity analysis showed that the decreased plasma exposure of active metabolites due to the decreased CES1 was partly attenuated by the decreased GFR. Conclusion: The developed PBPK model successfully predicted the pharmacokinetics of CES1 substrates and their metabolites in healthy individuals and LC patients, facilitating tailored dosing of CES1 substrates in LC patients.

1. Introduction

Liver cirrhosis (LC) is widely prevalent worldwide and results from a variety of causes including obesity, non-alcoholic fatty liver disease, high alcohol consumption, hepatitis B or C infection, autoimmune diseases, cholestatic diseases and iron or copper overload [1,2]. The Child–Pugh score is often used to classify liver cirrhosis into Child–Pugh A (CP-A), Child–Pugh B (CP-B) and Child–Pugh C (CP-C) according to the severity of LC [3,4]. In addition to the impairment of hepatic functions, LC also leads to remarkable alterations in a series of other physiological parameters such as functional liver volume, hepatic arterial blood flow, portal venous blood flow, glomerular filtration rate (GFR), α-acid glycoprotein, albumin content, drug-metabolizing enzymes and transporters. The alterations may directly affect the pharmacokinetics of drugs [5]. For example, Duthaler et al. investigated the effects of LC on the pharmacokinetics of CYP450 cocktail probes with caffeine (CYP1A2), efavirenz (CYP2B6), flurbiprofen (CYP2C9), omeprazole (CYP2C19), metoprolol (CYP2D6) and midazolam (CYP3A). They found that liver cirrhosis increased the plasma exposure of tested probes, the extent of which depended on the type of probe and LC severity. The calculated ratios of the AUC in patients to that in controls (AUCR) of caffeine, efavirenz, flurbiprofen, omeprazole, metoprolol and midazolam in CP-C patients were 6.2, 0.8, 1.4, 10.5, 4.5 and 6.3, respectively. The calculated AUCR values of omeprazole in CP-A, CP-B and CP-C patients were separately 4.8, 6.5 and 10.5. The AUCR values of probes in LC patients were in line with those in the contents of hepatic CYP450s [6]. LC also affects the renal excretion and intestinal absorption of drugs. Furosemide is primarily eliminated through the kidneys. It was reported [7] that clearance (CL) of furosemide significantly decreased from 154 mL/min in control subjects to 91 mL/min in CP-B or CP-C patients, which mainly resulted from decreases in renal clearance (CLK). These results indicate that drug dosage adjustments are necessary for LC patients based on the severity of their condition. Thus, regulatory agencies recommend pharmacokinetic studies of drugs in LC patients [8]. However, conducting pharmacokinetic studies in LC patients can be both costly and time-consuming. More importantly, it is difficult to recruit patients, especially patients with CP-C. Physiologically based pharmacokinetic (PBPK) modeling is considered an ideal technique for predicting the pharmacokinetics of drugs in patients with altered physiology. The alterations in physiological parameters, expression of hepatic drug-metabolizing enzymes and transporters under various degrees of severity of LC have been demonstrated. The possibilities for predicting the pharmacokinetics of drugs in LC patients using the PBPK model have been demonstrated [9].
Carboxylesterase1 (CES1) is one of the most abundant drug-metabolizing enzymes in human livers, constituting approximately 1% of the entire liver proteome. CES1 is responsible for 80–95% of total hydrolytic activity in the liver, which mediates the metabolism of a wide range of drugs, pesticides, environmental pollutants and endogenous compounds [10]. CES1-mediated metabolism leads to the biotransformation of a pharmacologically active drug into its inactive metabolite, as exemplified by methylphenidate hydrolysis. CES1 also mediates the activation of some prodrugs. The typical examples are some angiotensin-converting enzyme inhibitors (such as enalapril, cilazapril and temocapril) and neuraminidase inhibitors (oseltamivir). CES1 also hydrolyzes cholesteryl ester in lipid metabolism in human macrophages and hepatocytes, inferring that CES1 could be a potential drug target for the treatment of metabolic diseases, such as diabetes and atherosclerosis [10,11,12,13]. LC has been demonstrated to significantly downregulate expressions of the hepatic CES1 protein; the CES1 contents in CP-B patients and CP-C patients were decreased to 70% and 30% of those of healthy subjects, respectively, and the CES1 enzyme content in CP-A patients was comparable to that of healthy subjects [9]. On this basis, LC can alter the plasma exposure of its substrate drugs such as enalapril and oseltamivir [14,15]. Moreover, it is worth noting that metabolites of most CES1 substrates (such as enalapril and oseltamivir) are mainly eliminated via renal excretion. LC also injures renal functions, leading to decreases in renal clearance of the metabolites, indicating that alterations in the plasma exposure of metabolites by LC are attributed to the integrated effects of the decreases in hepatic CES1 activity and renal clearance.
This study aimed to develop a semi-PBPK model incorporating alterations in hepatic CES1 activity, liver/renal functions, gastrointestinal transit rate and relevant organ blood flow to simultaneously predict the pharmacokinetics of CES1 drugs and their metabolites in LC patients. Clinical pharmacokinetic studies of CES1 drugs were collected from data published on PubMed based on the following criteria. (1) The tested drug must be metabolized primarily by CES1. (2) Pharmacokinetic parameters (such as AUC or plasma drug concentrations) following intravenous (i.v.) and/or oral (p.o.) administration to liver cirrhosis populations must be available. (3) The clinical pharmacokinetic data might come from different reports. Based on these criteria, nine CES1 substrates were included in the simulations. The nine drugs are primarily metabolized by CES1 and include six prodrugs (enalapril, benazepril, cilazapril, perindopril, temocapril and oseltamivir) and three direct-acting drugs (flumazenil, pethidine and remimazolam). Flumazenil and remimazolam are mainly administered by intravenous injection. Pethidine is administrated via intravenous or oral routes. The remaining drugs are administered as oral immediate-release formulations. The predicted results were compared with clinical studies in patients with different statuses of LC. These results will assist in tailoring dosages of CES1 substrates in LC patients.

2. Materials and Methods

2.1. General Workflow

The workflow for developing a PBPK model (Figure 1) for LC patients. Initially, a semi-PBPK model (Figure 2) was developed for a virtual population of healthy individuals validated using clinical pharmacokinetic studies in healthy subjects. Then, the developed PBPK model was translated to LC patients by replacing the values of system-specific model parameters. Finally, pharmacokinetic predictions were conducted in 1000 virtual patients individuals and compared with clinical pharmacokinetic data from the literature.

2.2. Model Development

A semi-PBPK model was developed to simultaneously predict the pharmacokinetics of CES1 substrate drugs and their metabolites in LC patients. The semi-PBPK model consists of the stomach, intestinal wall, intestinal lumen, portal vein, liver, kidney and systemic compartment, which are connected by the blood circulatory system. The elimination of most drugs mainly occurs in the liver and kidneys. Drugs are administrated via the intravenous route or oral route. It is generally accepted that absorption of most orally administered drugs may occur in the small intestine (duodenum, jejunum and ileum). Absorbed amounts of drugs in the stomach, caecum and colon are minor. The effective permeability coefficient (Peff) is used to indicate the absorption capacity of a drug [16]. In the simulation, it was assumed that elimination of the tested drugs only occurred in the liver and kidneys and absorption of drugs only occurred in the small intestine.
All available information on anatomical, physiological and ADME parameters of the tested drugs was collected for the initial model construction (Table 1 and Table 2). Coding and solving of the PBPK model were conducted on WinNonlin 8.1 (Pharsight, St. Louis, MO, USA). The specific code and formulas for the model can be found in the Supplementary Material. After developing the initial model, parts of the plasma concentration curves of drugs from healthy subjects were used to estimate and optimize some parameters. Subsequently, the developed PBPK model was validated using plasma concentration–time curves from the rest of the clinical studies.

2.3. PBPK Model Development in LC Patients

The anatomical and physiological parameters in healthy subjects were replaced with those (Table 1) in LC patients. The LC-induced alterations in parameters related to ADME were estimated according to their values in healthy (HT) subjects and the altered physiological parameters.
For CES1-mediated hepatic metabolism,
C L i n t , C I , C E S 1 = C L i n t , H T , C E S 1 × f C E S 1 × f l i v e r
where CLint,CI,CES1 and CLint,HT,CES1 represent the values of CES1-mediated intrinsic clearance in the liver of patients and healthy subjects, respectively. fCES1 and fliver represent the ratio of CES1 content in patients to that in healthy subjects and liver volume in patients to that in healthy subjects, respectively.
For hepatic elimination of drugs mediated by other routes,
C L i n t , C I , o t h e r = C L i n t , H T × f o t h e r × f l i v e r
where CLint,cirr,other and CLint,heal,other represent the values of intrinsic clearance by other routes in the liver of patients and healthy subjects, respectively. fother is the ratio of other targets’ content in patients to that in healthy subjects.
Among the tested drugs, pethidine binds mainly to α1-acid glycoprotein and the rest bind mainly to albumin [88,93,94,95,96,97,98,99] (no data on binding protein for temocapril, so binding to albumin was assumed based on pka < 7.4, acidic). The free fraction of drugs in patient plasma was estimated using Equation (3) [21]:
f u , p , C I = 1 1 + ( 1 f u , p , H T ) × P p r o t , C I P p r o t , H T × f u , p , H T
where fu,p,CI, fu,p,HT, Pprot,CI and Pprot,HT represent the unbound fraction of the drug in the plasma of patients and healthy subjects and the concentration of drug-bound proteins in the plasma of patients and healthy subjects, respectively.
It was assumed that the free apparent volume of the distribution of the drug is unaltered; the apparent volume of distribution in cirrhosis patients (Vsys,CI) was derived from the apparent volume of distribution in healthy subjects, i,e.,
V s y s , C I = f u , p , C I f u , p , H T × V s y s , H T
Liver cirrhosis also impairs renal function and is characterized by a decrease in the glomerular filtration rate (GFR). The renal intrinsic clearance (CLint,K,CI) in patients may be estimated using equation [17]:
C L i n t , K , C I = C L i n t , K , H T × G F R C I / G F R H T
where CLint,k,HT, GFRHT and GFRCI represent renal intrinsic clearance in healthy subjects and GFR in healthy subjects and patients, respectively.
LC patients are often accompanied by impairment of the intestinal barrier [100]. The Lactulose/Rhamnose ratio is used to assess intestinal permeability [26]. The ratio of cirrhosis patients to healthy subjects was used to correct the absorption rate constant in LC patients:
P e f f , C I = P e f f , H T × L R C I / L R H T
where Peff,CI and Peff,HT are Peff values in LC patients and healthy subjects, respectively. LRCI and LRHT are, respectively, the Lactulose/Rhamnose ratios in LC patients and healthy subjects.
The four virtual populations (normal population, CP-A, CP-B and CP-C patients) were included in the simulations, each of which contained 1000 virtual individuals. For virtual population validation, each virtual individual was generated independently. CLint, CLint,K, fu,b, Vsystem, Peff, ka, KL:P, KG:P, and KK:P were used to generate virtual individuals. A random individual could be generated by taking random values in the range of 80–120% of the above parameter values. The 5th and 95th percentiles and average values of the simulation derived from 1000 virtual subjects were obtained. Effects of cirrhosis on the plasma exposure of the tested drugs were indexed as AUCR or CmaxR
A U C R = A U C C I A U C H T
Or
A U C R = C L H T C L C I
C m a x R = C m a x , C I C m a x , H T
where AUCCI, AUCHT, CLCI, CLHT, Cmax,CI and Cmax,HT are, respectively, the AUC, CL and Cmax of the tested drugs in cirrhosis patients and healthy subjects.

2.4. Criterion of the Developed PBPK Model

The PBPK model was considered to be successful if the simulated AUC or Cmax fell within 0.5- to 2-fold of the observed data or the observed data were within the 5th and 95th percentiles of the simulation derived from 1000 virtual subjects [101].

3. Results

3.1. Drug Data Set

Nine CES1 drugs, including six prodrugs (enalapril, benazepril, cilazapril, perindopril, temocapril and oseltamivir) and three direct-acting drugs (flumazenil, pethidine and remimazolam), were collected from data published on PubMed based on the following criteria. (1) The tested drug must be metabolized primarily by CES1. (2) Pharmacokinetic parameters (such as AUC or plasma drug concentrations) following intravenous (i.v.) and/or oral (p.o.) administration to liver cirrhosis populations must be available. (3) The clinical pharmacokinetic data might come from different reports. The collected pharmacokinetic parameters and drug information on clinical reports are listed in Table 2 and Table 3, respectively.

3.1.1. Enalapril and Enalaprilat

Enalapril, an angiotensin-converting enzyme inhibitor (ACEI), is a prodrug, which is mainly metabolized to the active product enalaprilat via hepatic CES1 [12,102]. Enalaprilat is eliminated primarily through the kidneys [103]. In plasma, enalapril and enalaprilat are mainly bound to albumin, and their free fractions in plasma are 0.55 and 0.5 [33]. Five clinical reports, including two reports involving liver cirrhosis, were selected in the simulations.
Table 3. Clinical information about CES1 substrates in the simulations.
Table 3. Clinical information about CES1 substrates in the simulations.
NoAuthorsDrugDose (mg)AnalytesSubjects (n)Ref
1Ohnishi A et al., 1989enalapril maleate10, p.oenalapril, enalaprilatHealthy (7)[14]
enalapril maleate10, p.oenalapril, enalaprilatCP-C (7)
2Todd PA et al., 1986enalapril maleate10, p.oenalapril, enalaprilatHealthy (12)[104]
3Weisser K et al., 1991enalapril maleate10, p.oenalapril, enalaprilatHealthy (8)[105]
4Dickstein K et al., 1987enalapril maleate10, p.oenalapril, enalaprilatHealthy (10)[106]
5Baba T et al., 1990enalapril maleate10, p.oenalapril, enalaprilatCP-B (7)[107]
6Kaiser G et al., 1989benazepril HCl10, p.obenazepril, benazeprilatHealthy (59)[108]
7Schweizer C et al., 1993benazepril HCl10, p.obenazepril, benazeprilatHealthy (11)[109]
8Sioufi A et al., 1994benazepril HCl20, p.obenazepril, benazeprilatHealthy (24)[110]
9Waldmeier F et al., 1991benazepril HCl20, p.obenazepril, benazeprilatHealthy (4)[111]
10Kaiser G et al., 1990benazepril HCl20, p.obenazepril, benazeprilatCP-B (12)[112]
11Macdonald NJ et al., 1993benazepril HCl10, p.obenazeprilatHealthy (18)[113]
12Massarella J et al., 1989cilazapril1.0, 2.5, 5, p.ocilazapril, cilazaprilatHealthy (24)[51]
13Williams PEO et al., 1990cilazapril2.5, p.ocilazapril, cilazaprilatHealthy (13)[114]
14Gross V et al., 1993cilazapril1, p.ocilazapril, cilazaprilatHealthy (10)[115]
cilazapril1, p.ocilazapril, cilazaprilatCP-B (9)
15Williams PEO et al., 1989cilazapril1, p.ocilazapril, cilazaprilatHealthy (12)[116]
16Massarella JW et al., 1989cilazapril5, p.ocilazapril, cilazaprilatHealthy (16)[117]
17Francis RJ et al., 1987cilazapril1.25, 2.5, 5,10, p.ocilazaprilatHealthy (12)[118]
18Lecocq B et al., 1990perindopril a4, p.operindopril, perindoprilatHealthy (12)[119]
19Tsai HH et al., 1989perindopril a8, p.operindopril, perindoprilatCP-A (8)[120]
20Thiollet M et al., 1992perindopril a8, p.operindopril, perindoprilatCP-B (10)[121]
21Lees KR et al., 1988perindopril a8, p.operindoprilatHealthy (8)[122]
22Furuta S et al., 1993temocapril HCl1, p.otemocapril, temocaprilatHealthy (6)[123]
temocapril HCl1, p.otemocapril, temocaprilatCP-C (7)
23Abe M et al., 2006oseltamivir b75, p.ooseltamivir,
oseltamivir carboxylate
Healthy (7)[124]
24Brewster M et al., 2006oseltamivir b75, p.ooseltamivir,
oseltamivir carboxylate
Healthy (18)[125]
25Jittamala P et al., 2014oseltamivir b75, p.ooseltamivir,
oseltamivir carboxylate
Healthy (12)[126]
oseltamivir b150, p.ooseltamivir,
oseltamivir carboxylate
Healthy (12)
26Snell P et al., 2005oseltamivir b75, p.ooseltamivir,
oseltamivir carboxylate
CP-B (11)[15]
27Amrei R et al., 1990flumazenil10 mg, i.v.flumazenilHealthy (NA)[127]
28Breimer LTM et al., 1991flumazenil10/10 min, ivflumazenilHealthy (7)[128]
29Pomier-Layrargues G
et al., 1989
flumazenil2/5 min, ivflumazenilCP-B (8)[129]
flumazenil2/5 min, ivflumazenilCP-C (8)
30Klotz U et al., 1984flumazenil2.5, i.vflumazenilHealthy (6)[81]
31Janssen U,et al., 1989flumazenil30, p.oflumazenilHealthy (8)[130]
flumazenil2, i.v; 30, p.oflumazenilCP-C (8)
32Verbeeck RK et al., 1981pethidine HCl25, i.vpethidineHealthy (6)[131]
pethidine HCl25, p.opethidineHealthy (6)
33Mather LE et al., 1975pethidine HCl50, i.vpethidineHealthy (4)[132]
34Kuhnert BR et al., 1980pethidine HCl50, i.vpethidineHealthy (7)[133]
35Guay DR et al., 1984pethidine HCl70, i.vpethidineHealthy (8)[134]
36Guay DR et al., 1985pethidine HCl70, i.vpethidineHealthy (8)[135]
37Pond SM et al., 1981pethidine HCl60, iv; 112, popethidineCP-A (5)[136]
38Pond SM et al., 1980pethidine HCl54.4, iv; 108.8, popethidineCP-B (4)[137]
39Mather LE et al., 1976pethidine HCl50, iv; 100, popethidineHealthy (4)[138]
40Klotz U et al., 1974pethidine HCl63.9, i.vpethidineHealthy (8)[139]
pethidine HCl53.1, i.vpethidineCP-A (10)
41Neal EA et al., 1979pethidine HCl56, iv; 56, popethidineHealthy (4)[140]
pethidine HCl56, iv; 56, popethidineCP-A (8)
42Sheng XY et al., 2020remimazolam besylate1.5425, 3.315, i.vremimazolamHealthy (3)[76]
remimazolam besylate4.8675, 6.18, i.vremimazolamHealthy (7)
remimazolam besylate13.26, 24.6, i.vremimazolamHealthy (8)
remimazolam besylate18.3, i.vremimazolamHealthy (10)
43Stohr T et al., 2021remimazolam besylate10.4, i.vremimazolamCP-B (8)[141]
remimazolam besylate8.2, i.vremimazolamCP-C (3)
a: Perindopril tert-butylamine; b: Oseltamivir phosphate.

3.1.2. Benazepril and Benazeprilat

Benazepril, a prodrug, is metabolized by hepatic CES1 to the active product benazeprilat [12,102], which shows inhibition of angiotensin-converting enzyme (ACE). Benazeprilat is eliminated via renal excretion. Benazepril and benazeprilat are mainly bound to albumin, belonging to drugs with high plasma binding, and their free fractions in plasma are 0.03 and 0.05 [47], respectively. Six clinical reports, including one report involving liver cirrhosis, were selected in the simulations.

3.1.3. Cilazapril and Cilazaprilat

Cilazapril is also metabolized by hepatic CES1 into cilazaprilat [12,102]. Cilazaprilat is mainly eliminated via the kidneys [52]. Cilazapril and cilazaprilat are mainly bound to albumin, belonging to medium plasma-binding drugs, and their free fractions in plasma are 0.70 and 0.76 [50], respectively. Six clinical reports, including one report involving liver cirrhosis, were selected in the simulations.

3.1.4. Perindopril and Perindoprilat

The prodrug perindopril is mainly metabolized by hepatic CES1 to perindoprilat, which shows inhibition of ACE. The bioavailability of perindopril is 66% [64]. Perindopril is primarily converted to perindoprilat in the liver, and other major metabolites of perindopril are perindopril glucuronide and perindopril lactam [142]. Since it is not clear which isoenzyme of UGT metabolizes perindopril to perindopril glucuronide, the change rate of AUC0-inf (0.62) for metoprolol in cirrhosis was used as a variation coefficient of intrinsic clearance for UGT [143]. Perindoprilat is eliminated via renal excretion. Perindopril and perindoprilat are predominantly bound to albumin. Perindopril shows higher plasma binding (percent binding 60%) than perindoprilat (mean percent binding 15%) [72].
Four clinical reports, including two reports involving liver cirrhosis, were selected in the simulations. Cirrhosis in perindopril and perindoprilat only have pharmacokinetic parameters and no specific drug concentration–time profile, so only a comparison of parameters was made.

3.1.5. Temocapril and Temocaprilat

Temocapril is also a prodrug and metabolized by hepatic CES1 to temocaprilat. Temocaprilat is eliminated via both bile and the kidneys. The biliary clearance of temocaprilat was about two times the renal clearance [65]. The CLint,K of temocaprilat was calculated to be 949.84 mL/min [64]. Thus, the CLbile,m of temocaprilat was estimated to be 1899.68 mL/min, assuming that the ratio of CLbile,m to CLint,K was 2.0. Biliary excretion of temocaprilat is considered to be mediated by multidrug resistance-associated protein2 (MRP2) [144]. One clinical report involving both liver cirrhosis patients and healthy subjects was selected in the simulations.

3.1.6. Oseltamivir and Oseltamivir Carboxylate

Oseltamivir, a prodrug, is metabolized via hepatic CES1 [12,102] to its active metabolite oseltamivir carboxylate (OC), which has an antiviral effect. About 80% of an orally administered dose of oseltamivir reaches the systemic circulation as the active metabolite. The absolute bioavailability of the active metabolite from orally administered oseltamivir is 75% [145]. About 60 to 70% of an oral oseltamivir dose appears in urine as the active metabolite and less than 5% as oseltamivir. Oseltamivir carboxylate is primarily eliminated via renal excretion, accounting for 93% of intravenous doses [38]. The CLint,K values of both oseltamivir and oseltamivir carboxylate exceed the GFR, indicating that renal elimination occurs via a combination of glomerular filtration and renal tubular secretion. Both oseltamivir and oseltamivir carboxylate are primarily bound to albumin; their bound fractions in plasma were approximately 42% and less than 3% [36]. Four clinical reports, including one report involving liver cirrhosis, were selected in the simulations.

3.1.7. Flumazenil

Flumazenil, a benzodiazepine receptor antagonist, is usually administered by intravenous injection [83]. Flumazenil is inactivated by hepatic CES1 to flumazenil acid and probably by CYP450-catalyzed N-dealkylation to N-demethylated flumazenil [146]. Flumazenil is predominantly bound to serum albumin, and its plasma protein binding is about 40% [85]. Five clinical reports, including two reports involving liver cirrhosis, were selected in the simulations.

3.1.8. Pethidine

Pethidine (meperidine) is a synthetic opioid commonly used for analgesia in humans. Pethidine is metabolized in the body by two different pathways [88,102]. The primary pathway is hepatic CES1 metabolism to pethidinic acid, an inactive metabolite. Another pathway is N-demethylation by CYP2B6 to normeperidine, a nonopioid active metabolite. The oral bioavailability of pethidine varies from 48–56% [147]. Pethidine was predominantly bound to α1-acid glycoprotein. In the simulation for healthy subjects, the free fraction of pethidine in plasma was 0.418 [88]. Ten clinical reports, including four reports involving liver cirrhosis, were selected in the simulations.

3.1.9. Remimazolam

Remimazolam, an ultrashort-acting sedative agent, is metabolized by hepatic CES1 to an inactive carboxy acid metabolite. The plasma protein binding of remimazolam is approximately 92%, predominantly serum albumin [77]. In the clinic, remimazolam is normally administered intravenously. Two clinical reports, including one report involving liver cirrhosis, were selected in the simulations.

3.2. Development of PBPK Model and Validation Using Pharmacokinetic Parameters from Healthy Subjects following i.v. or Oral Administrations

Plasma concentration–time profiles of the tested CES1 substrates and their active metabolites following i.v. or oral administration to healthy subjects were simulated using the developed PBPK model and compared with clinical observations. The results showed that most of the observed data of the tested agents fell within the 5th and 95th percentiles of the simulated data (Figure 3 and Figure S1). The corresponding pharmacokinetic parameters AUC, CL and Cmax were estimated using the mean of the simulated profiles derived from 1000 virtual individuals and compared with clinical observations (Table 4, Table 5, Table 6, Table 7, Table 8, Table 9, Table 10, Table 11 and Table 12). Most of the simulated pharmacokinetic parameter (AUC, CL and Cmax) values for all drugs were also within two-fold of observations (Table 4, Table 5, Table 6, Table 7, Table 8, Table 9, Table 10, Table 11 and Table 12 and Figure 4). All the results demonstrated that the PBPK model was successfully developed.

3.3. Prediction of Pharmacokinetic Profiles for CES1 Substrates and Their Active Metabolites following i.v. or Oral Administration to LC Patients Using the Developed PBPK Model

The developed PBPK model, following validation in healthy subjects, was used to predict the pharmacokinetic profiles of the selected CES1 substrates and their active metabolites following intravenous or oral administration to 1000 virtual LC patients (Figure 4), and their pharmacokinetic parameters were estimated using the mean pharmacokinetic profile derived from 1000 simulations (Table 4, Table 5, Table 6, Table 7, Table 8, Table 9, Table 10, Table 11 and Table 12). The results showed that except for oral pethidine, the majority of the drug concentrations in LC patients were well within the 5th and 95th percentiles of pharmacokinetic profiles derived from 1000 virtual LC patients. Most of the estimated pharmacokinetic parameters were also within 0.5–2.0-fold of observations (Figure 4), indicating that the developed PBPK model can predict alterations in pharmacokinetic behaviors of CES1 substrates and their metabolites in LC patients.
Extents of pharmacokinetic parameters under liver cirrhosis, AUCR and CmaxR were also predicted using the estimated pharmacokinetic parameters (Figure 5 and Figure 6). AUC or Cmax values may come from different clinical reports or different doses, thus, the AUC or Cmax values were normalized by dose and their mean values were used for estimating the AUCR or CmaxR. The results showed that the vast majority of the ratios of predicted AUCR and CmaxR are close to observed values, with only a few individual values differing significantly, indicating a good prediction. All these show that the PBPK model successfully predicted the pharmacokinetics of drugs in cirrhosis.

3.4. Sensitivity Analysis of Model Parameters

The plasma concentration–time curve of enalapril and enalaprilat following oral administration (10 mg) was used as an example for pharmacokinetic sensitivity. Some parameters such as gastrointestinal motility rate (Kt), intestinal absorption (Peff), hepatic arterial blood flow rates (QLA), portal vein blood flow rates (QPV), hepatic CES1 activity (CLint,L), kidney blood flow rates (QK), GFR, fu,b and fu,b,m (free fraction of metabolites in blood) may affect the pharmacokinetics of drugs and were selected for sensitivity analysis. According to the variations in the corresponding parameters listed in Table 1, the variations of QPV and QK were set to be 1/2-, 1- and 2-fold; QLA and CLint,L were 1/3-, 1- and 3-fold; variation in GFR was 0.5-, 1- and 1.5-fold; variation in fu,b was 0.7-, 1- and 1.3-fold for enalapril; and fu,b,m was 0.7-, 1- and 1.3-fold for enalaprilat. A report showed that Kt values under diabetic status were lower by about 2-fold compared to healthy subjects [148]. Table 1 also showed that Kt values under liver cirrhosis were about 1.3-fold those of healthy subjects. Here, variations of Kt were set to be 1/2-, 1- and 2-fold. Highly different Peff values of enalapril were reported [30,149,150]. For example, Thoms et al.’s reported Peff value of enalapril was 0.00125 cm/min [149], while the Peff value of enalapril reported by Chaturvedi et al. was 0.0127 cm/min [150]. Thus, variations in the Peff values of enalapril were set to be 1/3-, 1- and 3-fold. The results (Figure 7) show that these tested parameters affect the pharmacokinetics profile of drugs in varying degrees; their contributions to the AUC of enalapril were Peff > CLint,L > Kt > fu,b > QPV > GFR > QK > QLA and to that of enalaprilat were Peff > GFR > CLint,L > Kt > fu,b,m > QPV > QK > QLA. In addition to impairment of liver failure, LC patients were associated with increases in intestinal transit rates, intestinal permeability of drugs, QLA and fu,b (due to decreases in plasma-binding protein levels) and decreases in GFR, QK, CES1 activity and QPV, although increases in QL were reported in CP-C patients. The contributions of LC-induced alterations in Kt, QPV, CLint,L, Peff, GFR, QK and fu,b to the plasma concentrations of enalapril and enalaprilat following an oral dose of enalapril maleate (10 mg) administered to CP-C patients and their integrated effects were also simulated. The results showed that decreases in the CLint,L and increases in the Peff of enalapril increased plasma concentrations of enalapril, while the increases in fu,b and Kt and decreases in QPV obviously decreased plasma concentrations of enalapril following an oral dose of enalapril maleate; the net effects were an increase in the plasma concentrations of enalapril. For enalaprilat, increases in Peff and decreases in GFR, QK and QPV significantly increased the plasma concentration profiles of enalaprilat, while decreases in CES1 activity and increases in the Kt and fu,b,m of enalaprilat significantly decreased plasma concentrations following oral enalapril maleate administration. Their net effects were to decrease plasma concentrations of enalaprilat (Figure 7Q,R).

4. Discussion

Hepatic CES1 mediates the inactivation of direct-acting drugs or the activation of some prodrugs, most of whose active metabolites are mainly eliminated via the kidneys. In addition to hepatic dysfunction, LC is also associated with alterations in organ blood flow, decreases in plasma protein levels, increases in intestinal permeability of drugs and impairment of renal functions, commonly affecting the pharmacokinetics of CES1 substrate drugs and their metabolites. Both the whole-PBPK model and the semi-PBPK model have been widely applied to predict the pharmacokinetics of drugs, but compared with the whole-PBPK model, semi-PBPK model needs fewer parameters without losing key dynamic information [151], which may avoid overparameterization in the whole-PBPK model. Moreover, the semi-PBPK model may avoid some of the parameter estimation difficulties of whole-PBPK models [152]. The main contributions of the study were the successful development of a semi-PBPK model involving intestinal absorption, hepatic metabolism and renal excretion to simultaneously predict the pharmacokinetic profiles of nine CES1 substrates (six prodrugs and three direct-acting drugs) in both healthy subjects and LC patients. Most clinical observations were within the 5th and 95th percentiles of simulations derived from 1000 virtual subjects. Most of the estimated AUC and Cmax values were also within 0.5–2.0-fold of observations.
The extent of LC-induced alterations in the plasma exposure of CES1 substrates and their metabolites was also assessed using AUCR and CmaxR. It was found that although most of the clinically observed plasma concentrations for the tested agents were within the 5th and 95th percentiles of simulations, poorly predicted AUCR or CmaxR values were found in benazepril, temocaprilat, perindopril and perindoprilat. The predicted AUCR values of flumazenil and pethidine were lower than the clinical observations. Benazepril and temocaprilat belong to highly bound compounds, and their fu,b values were 0.03 and 0.025, respectively. In general, it is difficult to obtain an accurate plasma-binding measurement for highly bound compounds [153]. In addition to CES1, UGTs also mediate perindopril metabolism [142]. The isoenzyme of UGT involved in the metabolism of perindopril has not been identified. In the simulation, it was assumed that LC-induced alterations in the CLint, UGT of perindopril were similar to that of metoprolol [143]. LC patients with different etiologies show different amounts of hepatic CES1. In addition to CES1, other enzymes also mediate the metabolism of flumazenil [146]. Pethidine is co-metabolized by CES1 and CYP2B6 [88,102]. Several reports have demonstrated extensive interindividual variability in the expression of CYP2B6 [154] and CES1 [102]. All of these factors may be reasons leading to the differences between the predicted and the observed AUCR values, which need further investigation.
In general, LC-induced impairments of hepatic CES1 activity increase the plasma exposure of CES1 substrates, but sensitivity analysis revealed that the increases in the plasma concentrations of CES1 substrates in LC patients were only partially attributed to the impairment of hepatic CES1. Increases in the intestinal permeability of drugs were also observed in LC patients, contributing to increased plasma exposures of CES1 substrates. In contrast, LC-induced increases in intestinal transit rate and decreases in plasma-binding proteins and QPV obviously decreased the plasma exposure of CES1 substrates, which partly attenuated the increases in the plasma exposures of CES1 substrates caused by liver cirrhosis. Metabolites of the tested CES1 substrates are eliminated via the kidneys. The decreases in the plasma exposure of metabolites induced by the impairment of hepatic CES1 activity were also partly attenuated by LC-induced alterations in GFR and QK. Even under some conditions, levels of the metabolites are increased rather than decreased due to impaired renal function. For example, the AUC values of perindoprilat in CP-A and CP-B patients were obviously higher than those in healthy individuals; the observed AUCR values were 2.89 and 1.2, respectively, which were near to predictions (1.98 in CP-A patients and 2.04 in CP-B patients). These findings may partly explain clinical findings that although liver cirrhosis obviously increases the plasma levels of enalapril and perindopril, the magnitude of serum ACE-lowering effects by the two drugs was fairly comparable between LC patients and healthy subjects [14,120,121].
Plasma levels of the direct-acting drugs flumazenil, pethidine and remimazolam following their administration to LC patients were also successfully simulated. The observed AUCR values of remimazolam in LC patients could not be calculated due to a lack of observed pharmacokinetic parameters in LC patients, contrasting our expectation that the AUCR values in CP-B patients and CP-C patients would be 0.76 and 0.61, which may be explained by the fact that the increased plasma concentration by the impairment of hepatic CES1 may be attenuated by increases in hepatic arterial blood flow and increases in fu,b (Figure S2). The above simulations showed that the LC-induced impairments of hepatic CES1 activity may increase plasma levels of CES1 substrates (parent drug) and decrease plasma levels of their metabolites, if dosage adjustments are dependent on characteristics. For example, although LC obviously increases the plasma levels of enalapril and perindopril, the levels of enalaprilat and perindoprilat and the extent of decreases in serum ACE activity were obviously unaltered [14,120,121], indicating that no dosage adjustment of enalapril and perindopril in LC patients is required. The simulated levels of pethidine in the plasma of LC patients were higher than those in healthy subjects, which explained why the results in LC patients were consistent with the clinical observation that LC enhanced the CNS toxicity of pethidine [155], indicated that reduced dosages of pethidine in patients with hepatic insufficiency are needed [156].
However, this study also has some shortcomings. The predictions for healthy subjects were based on “ideal” healthy subjects (body weight assumed to be 70 kg) without considering gender, body weight, race and genetic variance of CES1. Genetic variation in CES1 also affects the pharmacokinetics of CES1 substrates [102]. During the simulation in LC patients, LC patients were considered “ideal” CP-A, CP-B or CP-C patients without considering LC etiology, gender and race. It was reported that the amount of CES1 protein in patients with hepatitis C cirrhosis was approximately 1.47-fold that of patients with alcoholic cirrhosis [157]. Similarly, it was reported that flumazenil might improve memory in patients with alcoholic cirrhosis but not in patients with nonalcoholic cirrhosis [158]. Moreover, the mean absolute CES1 protein expression in female livers was reported to be 17.3% higher than that in male livers [159].
LC patients are often accompanied by impairment of the intestinal barrier and renal function. LC may impair the intestinal barrier and renal function via various mechanisms [100,160]. The most common causes of LC are chronic liver diseases related to alcohol consumption, hepatitis virus infection, obesity and/or usage of drugs. Alcohol and drugs may directly impair the intestinal barrier. LC also leads to microbial alterations, which affect the intestinal epithelial barrier function directly or indirectly. For example, increased endotoxin levels directly downregulate the expression of intestinal tight junctions. Portal hypertension is a severe consequence of cirrhosis, which may lead to ascites, variceal hemorrhage and an impaired intestinal barrier [100]. LC may impair renal functions via activating the renin–angiotensin system, the sympathetic nervous system or nonosmotic hypersecretion of arginine vasopressin. Moreover, the translocation of bacteria and bacterial products from the intestinal lumen to the mesenteric lymph nodes stimulates inflammatory responses, increasing the production of proinflammatory cytokines. Moreover, the increased circulating levels of endotoxin or bacterial DNA also increase serum levels of cytokines, in turn, impairing renal function [160].

5. Conclusions

The developed PBPK model may successfully be applied simultaneously to predict the pharmacokinetics of CES1 substrate drugs and their active metabolites in healthy subjects and LC patients. The impact of physiological alteration under different degrees of LC on the pharmacokinetic behaviors of drugs may be accurately simulated. The simulated results will help in deciding whether the dosage of CES1 substrates should be adjusted for LC patients.

Supplementary Materials

The following supporting information can be downloaded at https://www.mdpi.com/article/10.3390/pharmaceutics16020234/s1, Figure S1: The observed (points) and predicted (lines) plasma concentrations of the tested CES1 substrates and their active metabolites following intravenous or oral administration to healthy subjects. Benazepril (A) and benazeprilat (B) following oral 10 mg benazepril hydrochloride; cilazapril (D,F) and cilazaprilat (C,E,G,H) following oral 1.25, 2.5, 5, 10 mg cilazapril; oseltamivir (I) and oseltamivir carboxylate (J) following oral 150 mg oseltamivir phosphate; flumazenil following intravenous 10 mg/1 min (K) and 10 mg/10 min (L); pethidine following intravenous 50 mg/1 min (M), 25 mg/1 min (N), 0.8 mg/kg, 1 min (O) and 0.8 mg/kg, 5 min (P); pethidine hydrochloride and oral 25 mg (Q), 0.8 mg/kg (R) pethidine hydrochloride; remimazolam following intravenous 0.05 (S), 0.075 (T), 0.2 (U), 0.3 (V), 0.4 (W) mg/kg remimazolam besylate. Shaded areas indicate the 5th and 95th percentiles of simulations derived from 1000 virtual individuals. The dashed lines indicate the mean of the simulated profiles. Figure S2: Contributions of LC-induced alterations in fu,b, CES1 activity, QLA and QPV to plasma concentrations of remimazolam following 10.4 mg (CP-B, A) and 8.2 mg (CP-C, B) administration to healthy human and LC patients.

Author Contributions

Conceptualization, X.L. (Xin Luo) and Z.Z.; methodology, X.L. (Xin Luo) and Z.Z.; validation, X.L. (Xin Luo), R.M. and G.H.; formal analysis, X.L. (Xin Luo) and Z.Z.; investigation, X.L. (Xin Luo) and G.H.; resources, X.L. (Xin Luo), R.M. and G.H.; data curation, X.L. (Xin Luo) and R.M.; writing—original draft preparation, X.L. (Xin Luo); writing—review and editing, L.L. and X.L. (Xiaodong Liu); supervision, L.L. and X.L. (Xiaodong Liu); project administration, L.L. and X.L. (Xiaodong Liu). All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the National Natural Science Foundation of China (No. 82073922 and 82173844) and the “Double First-Class” university project (No. CPU2022QZ21).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data are contained within the article and the Supplementary Materials.

Acknowledgments

The authors wish to thank Xiaodong Liu and Li Liu for their helpful advice in writing the English manuscript.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Gines, P.; Krag, A.; Abraldes, J.G.; Sola, E.; Fabrellas, N.; Kamath, P.S. Liver cirrhosis. Lancet 2021, 398, 1359–1376. [Google Scholar] [CrossRef] [PubMed]
  2. Lee, N.Y.; Suk, K.T. The Role of the Gut Microbiome in Liver Cirrhosis Treatment. Int. J. Mol. Sci. 2020, 22, 199. [Google Scholar] [CrossRef] [PubMed]
  3. El-Khateeb, E.; Darwich, A.S.; Achour, B.; Athwal, V.; Rostami-Hodjegan, A. Review article: Time to revisit Child-Pugh score as the basis for predicting drug clearance in hepatic impairment. Aliment. Pharmacol. Ther. 2021, 54, 388–401. [Google Scholar] [CrossRef] [PubMed]
  4. Pugh, R.N.; Murray-Lyon, I.M.; Dawson, J.L.; Pietroni, M.C.; Williams, R. Transection of the oesophagus for bleeding oesophageal varices. Br. J. Surg. 1973, 60, 646–649. [Google Scholar] [CrossRef] [PubMed]
  5. Weersink, R.A.; Burger, D.M.; Hayward, K.L.; Taxis, K.; Drenth, J.P.H.; Borgsteede, S.D. Safe use of medication in patients with cirrhosis: Pharmacokinetic and pharmacodynamic considerations. Expert Opin. Drug Metab. Toxicol. 2020, 16, 45–57. [Google Scholar] [CrossRef]
  6. Duthaler, U.; Bachmann, F.; Suenderhauf, C.; Grandinetti, T.; Pfefferkorn, F.; Haschke, M.; Hruz, P.; Bouitbir, J.; Krahenbuhl, S. Liver Cirrhosis Affects the Pharmacokinetics of the Six Substrates of the Basel Phenotyping Cocktail Differently. Clin. Pharmacokinet. 2022, 61, 1039–1055. [Google Scholar] [CrossRef]
  7. Villeneuve, J.P.; Verbeeck, R.K.; Wilkinson, G.R.; Branch, R.A. Furosemide kinetics and dynamics in patients with cirrhosis. Clin. Pharmacol. Ther. 1986, 40, 14–20. [Google Scholar] [CrossRef]
  8. Food and Drug Administration. Pharmacokinetics in Patients with Impaired Hepatic Function: Study Design, Data Analysis, and Impact on Dosing and Labeling. 2003. Available online: https://www.fda.gov/regulatory-information/search-fda-guidance-documents/pharmacokinetics-patients-impaired-hepatic-function-study-design-data-analysis-and-impact-dosing-and (accessed on 25 January 2024).
  9. Chen, Y.; Ke, M.; Xu, J.; Lin, C. Simulation of the Pharmacokinetics of Oseltamivir and Its Active Metabolite in Normal Populations and Patients with Hepatic Cirrhosis Using Physiologically Based Pharmacokinetic Modeling. AAPS PharmSciTech 2020, 21, 98. [Google Scholar] [CrossRef]
  10. Her, L.; Zhu, H.J. Carboxylesterase 1 and Precision Pharmacotherapy: Pharmacogenetics and Nongenetic Regulators. Drug Metab. Dispos. 2020, 48, 230–244. [Google Scholar] [CrossRef]
  11. Hosokawa, M. Structure and catalytic properties of carboxylesterase isozymes involved in metabolic activation of prodrugs. Molecules 2008, 13, 412–431. [Google Scholar] [CrossRef]
  12. Laizure, S.C.; Herring, V.; Hu, Z.; Witbrodt, K.; Parker, R.B. The role of human carboxylesterases in drug metabolism: Have we overlooked their importance? Pharmacotherapy 2013, 33, 210–222. [Google Scholar] [CrossRef] [PubMed]
  13. Ross, M.K.; Streit, T.M.; Herring, K.L. Carboxylesterases: Dual roles in lipid and pesticide metabolism. J. Pestic. Sci. 2010, 35, 257–264. [Google Scholar] [CrossRef] [PubMed]
  14. Ohnishi, A.; Tsuboi, Y.; Ishizaki, T.; Kubota, K.; Ohno, T.; Yoshida, H.; Kanezaki, A.; Tanaka, T. Kinetics and dynamics of enalapril in patients with liver cirrhosis. Clin. Pharmacol. Ther. 1989, 45, 657–665. [Google Scholar] [CrossRef] [PubMed]
  15. Snell, P.; Dave, N.; Wilson, K.; Rowell, L.; Weil, A.; Galitz, L.; Robson, R. Lack of effect of moderate hepatic impairment on the pharmacokinetics of oral oseltamivir and its metabolite oseltamivir carboxylate. Br. J. Clin. Pharmacol. 2005, 59, 598–601. [Google Scholar] [CrossRef]
  16. Gertz, M.; Harrison, A.; Houston, J.B.; Galetin, A. Prediction of human intestinal first-pass metabolism of 25 CYP3A substrates from in vitro clearance and permeability data. Drug Metab. Dispos. 2010, 38, 1147–1158. [Google Scholar] [CrossRef]
  17. Edginton, A.N.; Willmann, S. Physiology-based simulations of a pathological condition: Prediction of pharmacokinetics in patients with liver cirrhosis. Clin. Pharmacokinet. 2008, 47, 743–752. [Google Scholar] [CrossRef]
  18. Davies, B.; Morris, T. Physiological parameters in laboratory animals and humans. Pharm. Res. 1993, 10, 1093–1095. [Google Scholar] [CrossRef]
  19. Li, R.; Barton, H.A.; Maurer, T.S. A Mechanistic Pharmacokinetic Model for Liver Transporter Substrates Under Liver Cirrhosis Conditions. CPT Pharmacomet. Syst. Pharmacol. 2015, 4, 338–349. [Google Scholar] [CrossRef]
  20. Perdaems, N.; Blasco, H.; Vinson, C.; Chenel, M.; Whalley, S.; Cazade, F.; Bouzom, F. Predictions of metabolic drug-drug interactions using physiologically based modelling: Two cytochrome P450 3A4 substrates coadministered with ketoconazole or verapamil. Clin. Pharmacokinet. 2010, 49, 239–258. [Google Scholar] [CrossRef]
  21. Johnson, T.N.; Boussery, K.; Rowland-Yeo, K.; Tucker, G.T.; Rostami-Hodjegan, A. A semi-mechanistic model to predict the effects of liver cirrhosis on drug clearance. Clin. Pharmacokinet. 2010, 49, 189–206. [Google Scholar] [CrossRef]
  22. Gertz, M.; Houston, J.B.; Galetin, A. Physiologically based pharmacokinetic modeling of intestinal first-pass metabolism of CYP3A substrates with high intestinal extraction. Drug Metab. Dispos. 2011, 39, 1633–1642. [Google Scholar] [CrossRef] [PubMed]
  23. Badhan, R.; Penny, J.; Galetin, A.; Houston, J.B. Methodology for development of a physiological model incorporating CYP3A and P-glycoprotein for the prediction of intestinal drug absorption. J. Pharm. Sci. 2009, 98, 2180–2197. [Google Scholar] [CrossRef] [PubMed]
  24. Karlsen, S.; Fynne, L.; Gronbaek, H.; Krogh, K. Small intestinal transit in patients with liver cirrhosis and portal hypertension: A descriptive study. BMC Gastroenterol. 2012, 12, 176. [Google Scholar] [CrossRef] [PubMed]
  25. Rodriquez, A.; Martin, A.; Oterino, J.A.; Blanco, I.; Jimenez, M.; Perez, A.; Novoa, J.M. Renal function in compensated hepatic cirrhosis: Effects of an amino acid infusion and relationship with nitric acid. Dig. Dis. 1999, 17, 235–240. [Google Scholar] [CrossRef] [PubMed]
  26. Zuckerman, M.J.; Menzies, I.S.; Ho, H.; Gregory, G.G.; Casner, N.A.; Crane, R.S.; Hernandez, J.A. Assessment of intestinal permeability and absorption in cirrhotic patients with ascites using combined sugar probes. Dig. Dis. Sci. 2004, 49, 621–626. [Google Scholar] [CrossRef] [PubMed]
  27. Jacobsen, A.C.; Nielsen, S.; Brandl, M.; Bauer-Brandl, A. Drug Permeability Profiling Using the Novel Permeapad(R) 96-Well Plate. Pharm. Res. 2020, 37, 93. [Google Scholar] [CrossRef] [PubMed]
  28. Shin, B.S.; Yoon, C.H.; Balthasar, J.P.; Choi, B.Y.; Hong, S.H.; Kim, H.J.; Lee, J.B.; Hwang, S.W.; Yoo, S.D. Prediction of drug bioavailability in humans using immobilized artificial membrane phosphatidylcholine column chromatography and in vitro hepatic metabolic clearance. Biomed. Chromatogr. 2009, 23, 764–769. [Google Scholar] [CrossRef]
  29. Holford, N.H.G. Basic Principles. In Basic & Clinical Pharmacology, 12th ed.; Katzung, B.G., Masters, S.B., Trevor, A.J., Eds.; McGraw·Hill: New York City, NY, USA, 2012; p. 39. [Google Scholar]
  30. Dahlgren, D.; Roos, C.; Sjogren, E.; Lennernas, H. Direct In Vivo Human Intestinal Permeability (Peff) Determined with Different Clinical Perfusion and Intubation Methods. J. Pharm. Sci. 2015, 104, 2702–2726. [Google Scholar] [CrossRef]
  31. Tarkiainen, E.K.; Tornio, A.; Holmberg, M.T.; Launiainen, T.; Neuvonen, P.J.; Backman, J.T.; Niemi, M. Effect of carboxylesterase 1 c.428G > A single nucleotide variation on the pharmacokinetics of quinapril and enalapril. Br. J. Clin. Pharmacol. 2015, 80, 1131–1138. [Google Scholar] [CrossRef]
  32. Gangnus, T.; Burckhardt, B.B.; Consortium, C. Low-volume LC-MS/MS method for the pharmacokinetic investigation of carvedilol, enalapril and their metabolites in whole blood and plasma: Application to a paediatric clinical trial. Drug Test. Anal. 2021, 13, 694–708. [Google Scholar] [CrossRef]
  33. Claassen, K.; Willmann, S.; Eissing, T.; Preusser, T.; Block, M. A detailed physiologically based model to simulate the pharmacokinetics and hormonal pharmacodynamics of enalapril on the circulating endocrine Renin-Angiotensin-aldosterone system. Front. Physiol. 2013, 4, 4. [Google Scholar] [CrossRef] [PubMed]
  34. Faisal, M.; Cawello, W.; Burckhardt, B.B.; de Hoon, J.; Laer, S.; Consortium, L. Simultaneous Semi-Mechanistic Population Pharmacokinetic Modeling Analysis of Enalapril and Enalaprilat Serum and Urine Concentrations From Child Appropriate Orodispersible Minitablets. Front. Pediatr. 2019, 7, 281. [Google Scholar] [CrossRef] [PubMed]
  35. Hockings, N.; Ajayi, A.A.; Reid, J.L. Age and the pharmacokinetics of angiotensin converting enzyme inhibitors enalapril and enalaprilat. Br. J. Clin. Pharmacol. 1986, 21, 341–348. [Google Scholar] [CrossRef] [PubMed]
  36. Jogiraju, V.K.; Avvari, S.; Gollen, R.; Taft, D.R. Application of physiologically based pharmacokinetic modeling to predict drug disposition in pregnant populations. Biopharm. Drug Dispos. 2017, 38, 426–438. [Google Scholar] [CrossRef] [PubMed]
  37. Jhee, S.S.; Yen, M.; Ereshefsky, L.; Leibowitz, M.; Schulte, M.; Kaeser, B.; Boak, L.; Patel, A.; Hoffmann, G.; Prinssen, E.P.; et al. Low penetration of oseltamivir and its carboxylate into cerebrospinal fluid in healthy Japanese and Caucasian volunteers. Antimicrob. Agents Chemother. 2008, 52, 3687–3693. [Google Scholar] [CrossRef] [PubMed]
  38. He, G.; Massarella, J.; Ward, P. Clinical pharmacokinetics of the prodrug oseltamivir and its active metabolite Ro 64-0802. Clin. Pharmacokinet. 1999, 37, 471–484. [Google Scholar] [CrossRef] [PubMed]
  39. Snell, P.; Oo, C.; Dorr, A.; Barrett, J. Lack of pharmacokinetic interaction between the oral anti-influenza neuraminidase inhibitor prodrug oseltamivir and antacids. Br. J. Clin. Pharmacol. 2002, 54, 372–377. [Google Scholar] [CrossRef] [PubMed]
  40. Oh, J.; Lee, S.; Lee, H.; Cho, J.Y.; Yoon, S.H.; Jang, I.J.; Yu, K.S.; Lim, K.S. The novel carboxylesterase 1 variant c.662A>G may decrease the bioactivation of oseltamivir in humans. PLoS ONE 2017, 12, e0176320. [Google Scholar] [CrossRef]
  41. Hsueh, C.H.; Hsu, V.; Zhao, P.; Zhang, L.; Giacomini, K.M.; Huang, S.M. PBPK Modeling of the Effect of Reduced Kidney Function on the Pharmacokinetics of Drugs Excreted Renally by Organic Anion Transporters. Clin. Pharmacol. Ther. 2018, 103, 485–492. [Google Scholar] [CrossRef]
  42. Remko, M. Acidity, lipophilicity, solubility, absorption, and polar surface area of some ACE inhibitors. Chem. Pap. 2007, 61, 133–141. [Google Scholar] [CrossRef]
  43. Nishimuta, H.; Houston, J.B.; Galetin, A. Hepatic, intestinal, renal, and plasma hydrolysis of prodrugs in human, cynomolgus monkey, dog, and rat: Implications for in vitro-in vivo extrapolation of clearance of prodrugs. Drug Metab. Dispos. 2014, 42, 1522–1531. [Google Scholar] [CrossRef] [PubMed]
  44. Sun, J.X.; Cipriano, A.; Chan, K.; John, V.A. Pharmacokinetic interaction study between benazepril and amlodipine in healthy subjects. Eur. J. Clin. Pharmacol. 1994, 47, 285–289. [Google Scholar] [CrossRef] [PubMed]
  45. Wang, X.D.; Chan, E.; Chen, X.; Liao, X.X.; Tang, C.; Zhou, Z.W.; Huang, M.; Zhou, S.F. Simultaneous and rapid quantitation of benazepril and benazeprilat in human plasma by high performance liquid chromatography with ultraviolet detection. J. Pharm. Biomed. Anal. 2007, 44, 224–230. [Google Scholar] [CrossRef]
  46. Gatarić, B.B. Primena Tehnika za Naprednu Analizu Podataka u Biofarmaceutskoj Karakterizaciji Lekova: Identifikacija, Klasifikacija i Predviđanje Faktora Koji Utiču na Intestinalnu Apsorpciju Lekovitih Supstanci. Ph.D. Thesis, University of Belgrade, Belgrade, Serbia, 2021. [Google Scholar]
  47. Gengo, F.M.; Brady, E. The pharmacokinetics of benazepril relative to other ACE inhibitors. Clin. Cardiol. 1991, 14, IV44–IV55. [Google Scholar] [CrossRef] [PubMed]
  48. Chan, K.K.; Buch, A.; Glazer, R.D.; John, V.A.; Barr, W.H. Site-differential gastrointestinal absorption of benazepril hydrochloride in healthy volunteers. Pharm. Res. 1994, 11, 432–437. [Google Scholar] [CrossRef] [PubMed]
  49. Navia, M.; Chaturvedi, P. Design principles for orally bioavailable drugs. Drug Discov. Today 1996, 1, 179–189. [Google Scholar] [CrossRef]
  50. Wu, L.P.; Cui, Y.; Xiong, M.J.; Wang, S.R.; Chen, C.; Ye, L.M. Mixed micellar liquid chromatography methods: Modelling quantitative retention-activity relationships of angiotensin converting enzyme inhibitors. Biomed. Chromatogr. 2008, 22, 1243–1251. [Google Scholar] [CrossRef]
  51. Massarella, J.; DeFeo, T.; Lin, A.; Limjuco, R.; Brown, A. The pharmacokinetics and dose proportionality of cilazapril. Br. J. Clin. Pharmacol. 1989, 27 (Suppl. 2), 199S–204S. [Google Scholar] [CrossRef]
  52. Fillastre, J.P.; Moulin, B.; Godin, M.; Williams, P.E.; Brown, A.N.; Francis, R.J.; Pinta, P.; Manfredi, R. Pharmacokinetics of cilazapril in patients with renal failure. Br. J. Clin. Pharmacol. 1989, 27 (Suppl. 20), 275S–282S. [Google Scholar] [CrossRef]
  53. Kleinbloesem, C.H.; van Brummelen, P.; Francis, R.J.; Wiegand, U.W. Clinical pharmacology of cilazapril. Drugs 1991, 41 (Suppl. 1), 3–10. [Google Scholar] [CrossRef]
  54. Williams, P.E.; Brown, A.N.; Rajaguru, S.; Francis, R.J.; Walters, G.E.; McEwen, J.; Durnin, C. The pharmacokinetics and bioavailability of cilazapril in normal man. Br. J. Clin. Pharmacol. 1989, 27 (Suppl. 2), 181S–188S. [Google Scholar] [CrossRef]
  55. Sugihara, M.; Takeuchi, S.; Sugita, M.; Higaki, K.; Kataoka, M.; Yamashita, S. Analysis of Intra- and Intersubject Variability in Oral Drug Absorption in Human Bioequivalence Studies of 113 Generic Products. Mol. Pharm. 2015, 12, 4405–4413. [Google Scholar] [CrossRef] [PubMed]
  56. Kitagawa, S.; Takeda, J.; Sato, S. pH-dependent inhibitory effects of angiotensin-converting enzyme inhibitors on cefroxadine uptake by rabbit small intestinal brush-border membrane vesicles and their relationship with hydrophobicity and the ratio of zwitterionic species. Biol. Pharm. Bull. 1999, 22, 721–724. [Google Scholar] [CrossRef] [PubMed]
  57. Ohura, K. Evaluation of the Oral Absorption of Ester-type Prodrugs. Yakugaku Zasshi 2020, 140, 369–376. [Google Scholar] [CrossRef]
  58. Maeda, K.; Ieiri, I.; Yasuda, K.; Fujino, A.; Fujiwara, H.; Otsubo, K.; Hirano, M.; Watanabe, T.; Kitamura, Y.; Kusuhara, H.; et al. Effects of organic anion transporting polypeptide 1B1 haplotype on pharmacokinetics of pravastatin, valsartan, and temocapril. Clin. Pharmacol. Ther. 2006, 79, 427–439. [Google Scholar] [CrossRef] [PubMed]
  59. Puchler, K.; Eckl, K.M.; Fritsche, L.; Renneisen, K.; Neumayer, H.H.; Sierakowski, B.; Lavrijssen, A.T.; Thomsen, T.; Roots, I. Pharmacokinetics of temocapril and temocaprilat after 14 once daily oral doses of temocapril in hypertensive patients with varying degrees of renal impairment. Br. J. Clin. Pharmacol. 1997, 44, 531–536. [Google Scholar] [CrossRef] [PubMed]
  60. Shitara, Y.; Maeda, K.; Ikejiri, K.; Yoshida, K.; Horie, T.; Sugiyama, Y. Clinical significance of organic anion transporting polypeptides (OATPs) in drug disposition: Their roles in hepatic clearance and intestinal absorption. Biopharm. Drug Dispos. 2013, 34, 45–78. [Google Scholar] [CrossRef] [PubMed]
  61. Ohura, K.; Nozawa, T.; Murakami, K.; Imai, T. Evaluation of transport mechanism of prodrugs and parent drugs formed by intracellular metabolism in Caco-2 cells with modified carboxylesterase activity: Temocapril as a model case. J. Pharm. Sci. 2011, 100, 3985–3994. [Google Scholar] [CrossRef]
  62. Vistoli, G.; Pedretti, A.; Testa, B. Chemodiversity and molecular plasticity: Recognition processes as explored by property spaces. Future Med. Chem. 2011, 3, 995–1010. [Google Scholar] [CrossRef]
  63. Oguchi, H.; Miyasaka, M.; Koiwai, T.; Tokunaga, S.; Hora, K.; Sato, K.; Yoshie, T.; Shioya, H.; Furuta, S. Pharmacokinetics of temocapril and enalapril in patients with various degrees of renal insufficiency. Clin. Pharmacokinet. 1993, 24, 421–427. [Google Scholar] [CrossRef]
  64. Song, J.C.; White, C.M. Clinical pharmacokinetics and selective pharmacodynamics of new angiotensin converting enzyme inhibitors: An update. Clin. Pharmacokinet. 2002, 41, 207–224. [Google Scholar] [CrossRef] [PubMed]
  65. Suzuki, H.; Kawaratani, T.; Shioya, H.; Uji, Y.; Saruta, T. Study on pharmacokinetics of a new biliary excreted oral angiotensin converting enzyme inhibitor, temocapril (CS-622) in humans. Biopharm. Drug Dispos. 1993, 14, 41–50. [Google Scholar] [CrossRef] [PubMed]
  66. Helal, F.; Lane, M.E. Transdermal delivery of Angiotensin Converting Enzyme inhibitors. Eur. J. Pharm. Biopharm. 2014, 88, 1–7. [Google Scholar] [CrossRef] [PubMed]
  67. Ono, A.; Tomono, T.; Ogihara, T.; Terada, K.; Sugano, K. Investigation of biopharmaceutical drug properties suitable for orally disintegrating tablets. ADMET DMPK 2016, 4, 335–360. [Google Scholar] [CrossRef]
  68. Sun, H. Capture hydrolysis signals in the microsomal stability assay: Molecular mechanisms of the alkyl ester drug and prodrug metabolism. Bioorg. Med. Chem. Lett. 2012, 22, 989–995. [Google Scholar] [CrossRef] [PubMed]
  69. Hurst, M.; Jarvis, B. Perindopril: An updated review of its use in hypertension. Drugs 2001, 61, 867–896. [Google Scholar] [CrossRef] [PubMed]
  70. Devissaguet, J.P.; Ammoury, N.; Devissaguet, M.; Perret, L. Pharmacokinetics of perindopril and its metabolites in healthy volunteers. Fundam. Clin. Pharmacol. 1990, 4, 175–189. [Google Scholar] [CrossRef]
  71. Vrhovac, B.; Sarapa, N.; Bakran, I.; Huic, M.; Macolic-Sarinic, V.; Francetic, I.; Wolf-Coporda, A.; Plavsic, F. Pharmacokinetic changes in patients with oedema. Clin. Pharmacokinet. 1995, 28, 405–418. [Google Scholar] [CrossRef]
  72. Ghiadoni, L. Perindopril for the treatment of hypertension. Expert Opin. Pharmacother. 2011, 12, 1633–1642. [Google Scholar] [CrossRef]
  73. Li, Q.; Hao, Z.; Yu, Y.; Tang, Y. Bioequivalence study of two perindopril tert-butylamine tablet formulations in healthy Chinese subjects under fasting and fed conditions: A randomized, open-label, single-dose, crossover trial. Biomed. Pharmacother. 2021, 135, 111221. [Google Scholar] [CrossRef]
  74. Ogawa, R.; Stachnik, J.M.; Echizen, H. Clinical pharmacokinetics of drugs in patients with heart failure: An update (part 2, drugs administered orally). Clin. Pharmacokinet. 2014, 53, 1083–1114. [Google Scholar] [CrossRef] [PubMed]
  75. Zhou, J.; Curd, L.; Lohmer, L.L.; Ossig, J.; Schippers, F.; Stoehr, T.; Schmith, V. Population Pharmacokinetics of Remimazolam in Procedural Sedation with Nonhomogeneously Mixed Arterial and Venous Concentrations. Clin. Transl. Sci. 2021, 14, 326–334. [Google Scholar] [CrossRef] [PubMed]
  76. Sheng, X.Y.; Liang, Y.; Yang, X.Y.; Li, L.E.; Ye, X.; Zhao, X.; Cui, Y.M. Safety, pharmacokinetic and pharmacodynamic properties of single ascending dose and continuous infusion of remimazolam besylate in healthy Chinese volunteers. Eur. J. Clin. Pharmacol. 2020, 76, 383–391. [Google Scholar] [CrossRef] [PubMed]
  77. Kim, K.M. Remimazolam: Pharmacological characteristics and clinical applications in anesthesiology. Anesth. Pain Med. 2022, 17, 1–11. [Google Scholar] [CrossRef] [PubMed]
  78. Zhu, C.; Jiang, L.; Chen, T.M.; Hwang, K.K. A comparative study of artificial membrane permeability assay for high throughput profiling of drug absorption potential. Eur. J. Med. Chem. 2002, 37, 399–407. [Google Scholar] [CrossRef]
  79. Gottipati, G. Prediction of Human Systemic, Biologically Relevant Pharmacokinetic (PK) Properties Using Quantitative Structure Pharmacokinetic Relationships (QSPKR) and Interspecies Pharmacokinetic Allometric Scaling (PK-AS) Approaches for Four Different Pharmacological Classes of Compounds. Ph.D. Thesis, Virginia Commonwealth University, Richmond, VA, USA, 2014. [Google Scholar]
  80. Ellison, C.A. Structural and functional pharmacokinetic analogs for physiologically based pharmacokinetic (PBPK) model evaluation. Regul. Toxicol. Pharmacol. 2018, 99, 61–77. [Google Scholar] [CrossRef] [PubMed]
  81. Klotz, U.; Ziegler, G.; Reimann, I.W. Pharmacokinetics of the selective benzodiazepine antagonist Ro 15-1788 in man. Eur. J. Clin. Pharmacol. 1984, 27, 115–117. [Google Scholar] [CrossRef] [PubMed]
  82. Patel, R.D.; Kumar, S.P.; Patel, C.N.; Shankar, S.S.; Pandya, H.A.; Solanki, H.A. Parallel screening of drug-like natural compounds using Caco-2 cell permeability QSAR model with applicability domain, lipophilic ligand efficiency index and shape property: A case study of HIV-1 reverse transcriptase inhibitors. J. Mol. Struct. 2017, 1146, 80–95. [Google Scholar] [CrossRef]
  83. Karavokiros, K.A.; Tsipis, G.B. Flumazenil: A benzodiazepine antagonist. DICP 1990, 24, 976–981. [Google Scholar] [CrossRef]
  84. Paixao, P.; Gouveia, L.F.; Morais, J.A. Prediction of the in vitro intrinsic clearance determined in suspensions of human hepatocytes by using artificial neural networks. Eur. J. Pharm. Sci. 2010, 39, 310–321. [Google Scholar] [CrossRef]
  85. Klotz, U.; Kanto, J. Pharmacokinetics and clinical use of flumazenil (Ro 15-1788). Clin. Pharmacokinet. 1988, 14, 1–12. [Google Scholar] [CrossRef] [PubMed]
  86. Ghafourian, T.; Barzegar-Jalali, M.; Hakimiha, N.; Cronin, M.T. Quantitative structure-pharmacokinetic relationship modelling: Apparent volume of distribution. J. Pharm. Pharmacol. 2004, 56, 339–350. [Google Scholar] [CrossRef] [PubMed]
  87. Luttrell, W.E.; Castle, M.C. Species differences in the hydrolysis of meperidine and its inhibition by organophosphate compounds. Fundam. Appl. Toxicol. 1988, 11, 323–332. [Google Scholar] [CrossRef] [PubMed]
  88. Alsmadi, M.M.; Idkaidek, N. The Analysis of Pethidine Pharmacokinetics in Newborn Saliva, Plasma, and Brain Extracellular Fluid After Prenatal Intrauterine Exposure from Pregnant Mothers Receiving Intramuscular Dose Using PBPK Modeling. Eur. J. Drug Metab. Pharmacokinet. 2023, 48, 281–300. [Google Scholar] [CrossRef] [PubMed]
  89. Pond, S.M.; Kretschzmar, K.M. Effect of phenytoin on meperidine clearance and normeperidine formation. Clin. Pharmacol. Ther. 1981, 30, 680–686. [Google Scholar] [CrossRef] [PubMed]
  90. Chan, K.; Tse, J.; Jennings, F.; Orme, M.L. Pharmacokinetics of low-dose intravenous pethidine in patients with renal dysfunction. J. Clin. Pharmacol. 1987, 27, 516–522. [Google Scholar] [CrossRef] [PubMed]
  91. Paixao, P.; Gouveia, L.F.; Morais, J.A. Prediction of the human oral bioavailability by using in vitro and in silico drug related parameters in a physiologically based absorption model. Int. J. Pharm. 2012, 429, 84–98. [Google Scholar] [CrossRef] [PubMed]
  92. Piscitelli, S.C.; Kress, D.R.; Bertz, R.J.; Pau, A.; Davey, R. The effect of ritonavir on the pharmacokinetics of meperidine and normeperidine. Pharmacotherapy 2000, 20, 549–553. [Google Scholar] [CrossRef]
  93. Toutain, P.L.; Lefebvre, H.P.; King, J.N. Benazeprilat disposition and effect in dogs revisited with a pharmacokinetic/pharmacodynamic modeling approach. J. Pharmacol. Exp. Ther. 2000, 280, 1087–1093. [Google Scholar]
  94. Pan, D.Q.; Jiang, M.; Liu, T.T.; Wang, Q.; Shi, J.H. Combined spectroscopies and molecular docking approach to characterizing the binding interaction of enalapril with bovine serum albumin. Luminescence 2017, 32, 481–490. [Google Scholar] [CrossRef]
  95. Lee, A.; Shirley, M. Remimazolam: A Review in Procedural Sedation. Drugs 2021, 81, 1193–1201. [Google Scholar] [CrossRef] [PubMed]
  96. Blei, A.T. Albumin dialysis for the treatment of hepatic encephalopathy. J. Gastroenterol. Hepatol. 2004, 19, S224–S228. [Google Scholar] [CrossRef]
  97. Nafisi, S.; Vishkaee, T.S. Study on the interaction of tamiflu and oseltamivir carboxylate with human serum albumin. J. Photochem. Photobiol. B 2011, 105, 34–39. [Google Scholar] [CrossRef] [PubMed]
  98. Obradovic, D.; Radan, M.; Dikic, T.; Nikolic, M.P.; Oljacic, S.; Nikolic, K. The evaluation of drug-plasma protein binding interaction on immobilized human serum albumin stationary phase, aided by different computational approaches. J. Pharm. Biomed. Anal. 2022, 211, 114593. [Google Scholar] [CrossRef] [PubMed]
  99. Anderson, P.J.; Critchley, J.A.; Tomlinson, B.; Resplandy, G. Comparison of the pharmacokinetics and pharmacodynamics of oral doses of perindopril in normotensive Chinese and Caucasian volunteers. Br. J. Clin. Pharmacol. 1995, 39, 361–368. [Google Scholar] [CrossRef] [PubMed]
  100. Pijls, K.E.; Jonkers, D.M.; Elamin, E.E.; Masclee, A.A.; Koek, G.H. Intestinal epithelial barrier function in liver cirrhosis: An extensive review of the literature. Liver Int. 2013, 33, 1457–1469. [Google Scholar] [CrossRef] [PubMed]
  101. Ladumor, M.K.; Storelli, F.; Liang, X.; Lai, Y.; Enogieru, O.J.; Chothe, P.P.; Evers, R.; Unadkat, J.D. Predicting changes in the pharmacokinetics of CYP3A-metabolized drugs in hepatic impairment and insights into factors driving these changes. CPT Pharmacomet. Syst. Pharmacol. 2023, 12, 261–273. [Google Scholar] [CrossRef]
  102. Chen, F.; Zhang, B.; Parker, R.B.; Laizure, S.C. Clinical implications of genetic variation in carboxylesterase drug metabolism. Expert Opin. Drug Metab. Toxicol. 2018, 14, 131–142. [Google Scholar] [CrossRef]
  103. Gomez, H.J.; Cirillo, V.J.; Irvin, J.D. Enalapril: A review of human pharmacology. Drugs 1985, 30 (Suppl. 1), 13–24. [Google Scholar] [CrossRef]
  104. Todd, P.A.; Heel, R.C. Enalapril. A review of its pharmacodynamic and pharmacokinetic properties, and therapeutic use in hypertension and congestive heart failure. Drugs 1986, 31, 198–248. [Google Scholar] [CrossRef]
  105. Weisser, K.; Schloos, J.; Lehmann, K.; Dusing, R.; Vetter, H.; Mutschler, E. Pharmacokinetics and converting enzyme inhibition after morning and evening administration of oral enalapril to healthy subjects. Eur. J. Clin. Pharmacol. 1991, 40, 95–99. [Google Scholar] [CrossRef]
  106. Dickstein, K.; Till, A.E.; Aarsland, T.; Tjelta, K.; Abrahamsen, A.M.; Kristianson, K.; Gomez, H.J.; Gregg, H.; Hichens, M. The pharmacokinetics of enalapril in hospitalized patients with congestive heart failure. Br. J. Clin. Pharmacol. 1987, 23, 403–410. [Google Scholar] [CrossRef] [PubMed]
  107. Baba, T.; Murabayashi, S.; Tomiyama, T.; Takebe, K. The pharmacokinetics of enalapril in patients with compensated liver cirrhosis. Br. J. Clin. Pharmacol. 1990, 29, 766–769. [Google Scholar] [CrossRef] [PubMed]
  108. Kaiser, G.; Ackermann, R.; Brechbuhler, S.; Dieterle, W. Pharmacokinetics of the angiotensin converting enzyme inhibitor benazepril.HCl (CGS 14 824 A) in healthy volunteers after single and repeated administration. Biopharm. Drug Dispos. 1989, 10, 365–376. [Google Scholar] [CrossRef] [PubMed]
  109. Schweizer, C.; Kaiser, G.; Dieterle, W.; Mann, J. Pharmacokinetics and pharmacodynamics of benazepril hydrochloride in patients with major proteinuria. Eur. J. Clin. Pharmacol. 1993, 44, 463–466. [Google Scholar] [CrossRef] [PubMed]
  110. Sioufi, A.; Pommier, F.; Gauducheau, N.; Godbillon, J.; Choi, L.; John, V. The absence of a pharmacokinetic interaction between aspirin and the angiotensin-converting enzyme inhibitor benazepril in healthy volunteers. Biopharm. Drug Dispos. 1994, 15, 451–461. [Google Scholar] [CrossRef]
  111. Waldmeier, F.; Kaiser, G.; Ackermann, R.; Faigle, J.W.; Wagner, J.; Barner, A.; Lasseter, K.C. The disposition of [14C]-labelled benazepril HCl in normal adult volunteers after single and repeated oral dose. Xenobiotica 1991, 21, 251–261. [Google Scholar] [CrossRef] [PubMed]
  112. Kaiser, G.; Ackermann, R.; Gschwind, H.P.; James, I.M.; Sprengers, D.; McIntyre, N.; Defalco, A.; Holmes, I.B. The influence of hepatic cirrhosis on the pharmacokinetics of benazepril hydrochloride. Biopharm. Drug Dispos. 1990, 11, 753–764. [Google Scholar] [CrossRef] [PubMed]
  113. Macdonald, N.J.; Sioufi, A.; Howie, C.A.; Wade, J.R.; Elliott, H.L. The effects of age on the pharmacokinetics and pharmacodynamics of single oral doses of benazepril and enalapril. Br. J. Clin. Pharmacol. 1993, 36, 205–209. [Google Scholar] [CrossRef]
  114. Williams, P.E.; Brown, A.N.; Rajaguru, S.; Francis, R.J.; Bell, A.J.; Dewland, P.M. Pharmacokinetics of cilazapril during repeated oral dosing in healthy young volunteers. Eur. J. Drug Metab. Pharmacokinet. 1990, 15, 63–67. [Google Scholar] [CrossRef]
  115. Gross, V.; Treher, E.; Haag, K.; Neis, W.; Wiegand, U.; Scholmerich, J. Angiotensin-converting enzyme (ACE)-inhibition in cirrhosis. Pharmacokinetics and dynamics of the ACE-inhibitor cilazapril (Ro 31-2848). J. Hepatol. 1993, 17, 40–47. [Google Scholar] [CrossRef] [PubMed]
  116. Williams, P.E.; Brown, A.N.; Rajaguru, S.; Walters, G.E.; McEwen, J.; Durnin, C. A pharmacokinetic study of cilazapril in elderly and young volunteers. Br. J. Clin. Pharmacol. 1989, 27 (Suppl. 2), 211S–215S. [Google Scholar] [CrossRef] [PubMed]
  117. Massarella, J.W.; DeFeo, T.M.; Brown, A.N.; Lin, A.; Wills, R.J. The influence of food on the pharmacokinetics and ACE inhibition of cilazapril. Br. J. Clin. Pharmacol. 1989, 27 (Suppl. 2), 205S–209S. [Google Scholar] [CrossRef] [PubMed]
  118. Francis, R.J.; Brown, A.N.; Kler, L.; Fasanella d’Amore, T.; Nussberger, J.; Waeber, B.; Brunner, H.R. Pharmacokinetics of the converting enzyme inhibitor cilazapril in normal volunteers and the relationship to enzyme inhibition: Development of a mathematical model. J. Cardiovasc. Pharmacol. 1987, 9, 32–38. [Google Scholar] [CrossRef] [PubMed]
  119. Lecocq, B.; Funck-Brentano, C.; Lecocq, V.; Ferry, A.; Gardin, M.E.; Devissaguet, M.; Jaillon, P. Influence of food on the pharmacokinetics of perindopril and the time course of angiotensin-converting enzyme inhibition in serum. Clin. Pharmacol. Ther. 1990, 47, 397–402. [Google Scholar] [CrossRef] [PubMed]
  120. Tsai, H.H.; Lees, K.R.; Howden, C.W.; Reid, J.L. The pharmacokinetics and pharmacodynamics of perindopril in patients with hepatic cirrhosis. Br. J. Clin. Pharmacol. 1989, 28, 53–59. [Google Scholar] [CrossRef]
  121. Thiollet, M.; Funck-Brentano, C.; Grange, J.D.; Midavaine, M.; Resplandy, G.; Jaillon, P. The pharmacokinetics of perindopril in patients with liver cirrhosis. Br. J. Clin. Pharmacol. 1992, 33, 326–328. [Google Scholar] [CrossRef]
  122. Lees, K.R.; Green, S.T.; Reid, J.L. Influence of age on the pharmacokinetics and pharmacodynamics of perindopril. Clin. Pharmacol. Ther. 1988, 44, 418–425. [Google Scholar] [CrossRef]
  123. Furuta, S.; Kiyosawa, K.; Higuchi, M.; Kasahara, H.; Saito, H.; Shioya, H.; Oguchi, H. Pharmacokinetics of temocapril, an ACE inhibitor with preferential biliary excretion, in patients with impaired liver function. Eur. J. Clin. Pharmacol. 1993, 44, 383–385. [Google Scholar] [CrossRef]
  124. Abe, M.; Smith, J.; Urae, A.; Barrett, J.; Kinoshita, H.; Rayner, C.R. Pharmacokinetics of oseltamivir in young and very elderly subjects. Ann. Pharmacother. 2006, 40, 1724–1730. [Google Scholar] [CrossRef]
  125. Brewster, M.; Smith, J.R.; Dutkowski, R.; Robson, R. Active metabolite from Tamiflu solution is bioequivalent to that from capsule delivery in healthy volunteers: A cross-over, randomised, open-label study. Vaccine 2006, 24, 6660–6663. [Google Scholar] [CrossRef] [PubMed]
  126. Jittamala, P.; Pukrittayakamee, S.; Tarning, J.; Lindegardh, N.; Hanpithakpong, W.; Taylor, W.R.; Lawpoolsri, S.; Charunwattana, P.; Panapipat, S.; White, N.J.; et al. Pharmacokinetics of orally administered oseltamivir in healthy obese and nonobese Thai subjects. Antimicrob. Agents Chemother. 2014, 58, 1615–1621. [Google Scholar] [CrossRef] [PubMed]
  127. Amrein, R.; Hetzel, W. Pharmacology of Dormicum (Midazolam) and Anexate (Flumazenil). Acta Anaesth. Scand. 1990, 34, 6–15. [Google Scholar] [CrossRef]
  128. Breimer, L.T.; Hennis, P.J.; Burm, A.G.; Danhof, M.; Bovill, J.G.; Spierdijk, J.; Vletter, A.A. Pharmacokinetics and EEG effects of flumazenil in volunteers. Clin. Pharmacokinet. 1991, 20, 491–496. [Google Scholar] [CrossRef] [PubMed]
  129. Pomier-Layrargues, G.; Giguere, J.F.; Lavoie, J.; Willems, B.; Butterworth, R.F. Pharmacokinetics of benzodiazepine antagonist Ro 15-1788 in cirrhotic patients with moderate or severe liver dysfunction. Hepatology 1989, 10, 969–972. [Google Scholar] [CrossRef] [PubMed]
  130. Janssen, U.; Walker, S.; Maier, K.; von Gaisberg, U.; Klotz, U. Flumazenil disposition and elimination in cirrhosis. Clin. Pharmacol. Ther. 1989, 46, 317–323. [Google Scholar] [CrossRef]
  131. Verbeeck, R.K.; Branch, R.A.; Wilkinson, G.R. Meperidine disposition in man: Influence of urinary pH and route of administration. Clin. Pharmacol. Ther. 1981, 30, 619–628. [Google Scholar] [CrossRef]
  132. Mather, L.E.; Tucker, G.T.; Pflug, A.E.; Lindop, M.J.; Wilkerson, C. Meperidine kinetics in man. Intravenous injection in surgical patients and volunteers. Clin. Pharmacol. Ther. 1975, 17, 21–30. [Google Scholar] [CrossRef]
  133. Kuhnert, B.R.; Kuhnert, P.M.; Prochaska, A.L.; Sokol, R.J. Meperidine disposition in mother, neonate, and nonpregnant females. Clin. Pharmacol. Ther. 1980, 27, 486–491. [Google Scholar] [CrossRef]
  134. Guay, D.R.; Meatherall, R.C.; Chalmers, J.L.; Grahame, G.R. Cimetidine alters pethidine disposition in man. Br. J. Clin. Pharmacol. 1984, 18, 907–914. [Google Scholar] [CrossRef]
  135. Guay, D.R.; Meatherall, R.C.; Chalmers, J.L.; Grahame, G.R.; Hudson, R.J. Ranitidine does not alter pethidine disposition in man. Br. J. Clin. Pharmacol. 1985, 20, 55–59. [Google Scholar] [CrossRef]
  136. Pond, S.M.; Tong, T.; Benowitz, N.L.; Jacob, P.; Rigod, J. Presystemic metabolism of meperidine to normeperidine in normal and cirrhotic subjects. Clin. Pharmacol. Ther. 1981, 30, 183–188. [Google Scholar] [CrossRef] [PubMed]
  137. Pond, S.M.; Tong, T.; Benowitz, N.L.; Jacob, P. Enhanced bioavailability of pethidine and pentazocine in patients with cirrhosis of the liver. Aust. N. Z. J. Med. 1980, 10, 515–519. [Google Scholar] [CrossRef] [PubMed]
  138. Mather, L.E.; Tucker, G.T. Systemic availability of orally administered meperidine. Clin. Pharmacol. Ther. 1976, 20, 535–540. [Google Scholar] [CrossRef] [PubMed]
  139. Klotz, U.; McHorse, T.S.; Wilkinson, G.R.; Schenker, S. The effect of cirrhosis on the disposition and elimination of meperidine in man. Clin. Pharmacol. Ther. 1974, 16, 667–675. [Google Scholar] [CrossRef] [PubMed]
  140. Neal, E.A.; Meffin, P.J.; Gregory, P.B.; Blaschke, T.F. Enhanced Bioavailability and Decreased Clearance of Analgesics in Patients with Cirrhosis. Gastroenterology 1979, 77, 96–102. [Google Scholar] [CrossRef] [PubMed]
  141. Stohr, T.; Colin, P.J.; Ossig, J.; Pesic, M.; Borkett, K.; Winkle, P.; Struys, M.; Schippers, F. Pharmacokinetic properties of remimazolam in subjects with hepatic or renal impairment. Br. J. Anaesth. 2021, 127, 415–423. [Google Scholar] [CrossRef] [PubMed]
  142. Grislain, L.; Mocquard, M.T.; Dabe, J.F.; Bertrand, M.; Luijten, W.; Marchand, B.; Resplandy, G.; Devissaguet, M. Interspecies comparison of the metabolic pathways of perindopril, a new angiotensin-converting enzyme (ACE) inhibitor. Xenobiotica 1990, 20, 787–800. [Google Scholar] [CrossRef] [PubMed]
  143. Duthaler, U.; Bachmann, F.; Ozbey, A.C.; Umehara, K.; Parrott, N.; Fowler, S.; Krahenbuhl, S. The Activity of Members of the UDP-Glucuronosyltransferase Subfamilies UGT1A and UGT2B is Impaired in Patients with Liver Cirrhosis. Clin. Pharmacokinet. 2023, 62, 1141–1155. [Google Scholar] [CrossRef]
  144. Ishizuka, H.; Konno, K.; Naganuma, H.; Sasahara, K.; Kawahara, Y.; Niinuma, K.; Suzuki, H.; Sugiyama, Y. Temocaprilat, a novel angiotensin-converting enzyme inhibitor, is excreted in bile via an ATP-dependent active transporter (cMOAT) that is deficient in Eisai hyperbilirubinemic mutant rats (EHBR). J. Pharmacol. Exp. Ther. 1997, 280, 1304–1311. [Google Scholar]
  145. Gao, G.; Law, F.; Wong, R.N.S.; Mak, N.K.; Yang, M.S.M. A physiologically-based pharmacokinetic model of oseltamivir phosphate and its carboxylate metabolite for rats and humans. ADMET DMPK 2019, 7, 22–43. [Google Scholar] [CrossRef]
  146. Kleingeist, B.; Bocker, R.; Geisslinger, G.; Brugger, R. Isolation and pharmacological characterization of microsomal human liver flumazenil carboxylesterase. J. Pharm. Pharm. Sci. A Publ. Can. Soc. Pharm. Sci. Soc. Can. Des. Sci. Pharm. 1998, 1, 38–46. [Google Scholar]
  147. Tegeder, I.; Lötsch, J.; Geisslinger, G. Pharmacokinetics of Opioids in Liver Disease. Clin. Pharmacokinet. 1999, 37, 17–40. [Google Scholar] [CrossRef] [PubMed]
  148. Iida, M.; Ikeda, M.; Kishimoto, M.; Tsujino, T.; Kaneto, H.; Matsuhisa, M.; Kajimoto, Y.; Watarai, T.; Yamasaki, Y.; Hori, M. Evaluation of gut motility in type II diabetes by the radiopaque marker method. J. Gastroenterol. Hepatol. 2000, 15, 381–385. [Google Scholar] [CrossRef] [PubMed]
  149. Thomas, S.; Brightman, F.; Gill, H.; Lee, S.; Pufong, B. Simulation modelling of human intestinal absorption using Caco-2 permeability and kinetic solubility data for early drug discovery. J. Pharm. Sci. 2008, 97, 4557–4574. [Google Scholar] [CrossRef]
  150. Chaturvedi, P.R.; Decker, C.J.; Odinecs, A. Prediction of pharmacokinetic properties using experimental approaches during early drug discovery. Curr. Opin. Chem. Biol. 2001, 5, 452–463. [Google Scholar] [CrossRef] [PubMed]
  151. Okino, M.S.; Mavrovouniotis, M.L. Simplification of Mathematical Models of Chemical Reaction Systems. Chem. Rev. 1998, 98, 391–408. [Google Scholar] [CrossRef]
  152. Tsamandouras, N.; Rostami-Hodjegan, A.; Aarons, L. Combining the ‘bottom up’ and ‘top down’ approaches in pharmacokinetic modelling: Fitting PBPK models to observed clinical data. Br. J. Clin. Pharmacol. 2014, 79, 48–55. [Google Scholar] [CrossRef]
  153. Riccardi, K.; Cawley, S.; Yates, P.D.; Chang, C.; Funk, C.; Niosi, M.; Lin, J.; Di, L. Plasma Protein Binding of Challenging Compounds. J. Pharm. Sci. 2015, 104, 2627–2636. [Google Scholar] [CrossRef]
  154. Turpeinen, M.; Zanger, U.M. Cytochrome P450 2B6: Function, genetics, and clinical relevance. Drug Metab. Drug Interact. 2012, 27, 185–197. [Google Scholar] [CrossRef]
  155. Danziger, L.H.; Martin, S.J.; Blum, R.A. Central Nervous System Toxicity Associated with Meperidine Use in Hepatic Disease. Pharmacother. J. Hum. Pharmacol. Drug Ther. 1994, 14, 235–238. [Google Scholar] [CrossRef]
  156. Soleimanpour, H.; Safari, S.; Shahsavari Nia, K.; Sanaie, S.; Alavian, S.M. Opioid Drugs in Patients with Liver Disease: A Systematic Review. Hepat. Mon. 2016, 16, e32636. [Google Scholar] [CrossRef]
  157. Prasad, B.; Bhatt, D.K.; Johnson, K.; Chapa, R.; Chu, X.; Salphati, L.; Xiao, G.; Lee, C.; Hop, C.; Mathias, A.; et al. Abundance of Phase 1 and 2 Drug-Metabolizing Enzymes in Alcoholic and Hepatitis C Cirrhotic Livers: A Quantitative Targeted Proteomics Study. Drug Metab. Dispos. 2018, 46, 943–952. [Google Scholar] [CrossRef]
  158. Kapczinski, F.; Sherman, D.; Williams, R.; Lader, M.; Curran, V. Differential effects of flumazenil in alcoholic and nonalcoholic cirrhotic patients. Psychopharmacology 1995, 120, 220–226. [Google Scholar] [CrossRef]
  159. Shi, J.; Wang, X.; Nguyen, J.H.; Bleske, B.E.; Liang, Y.; Liu, L.; Zhu, H.J. Dabigatran etexilate activation is affected by the CES1 genetic polymorphism G143E (rs71647871) and gender. Biochem. Pharmacol. 2016, 119, 76–84. [Google Scholar] [CrossRef]
  160. Gines, P.; Schrier, R.W. Renal failure in cirrhosis. N. Engl. J. Med. 2009, 361, 1279–1290. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Workflow for developing a semi-PBPK model. It involved establishing a PBPK model in normal subjects and validating it with a virtual population. Afterward, the parameters were changed according to the effects of cirrhosis and a model of PBPK in cirrhosis patients was created. Simulations were performed in virtual populations and compared with clinical pharmacokinetic data.
Figure 1. Workflow for developing a semi-PBPK model. It involved establishing a PBPK model in normal subjects and validating it with a virtual population. Afterward, the parameters were changed according to the effects of cirrhosis and a model of PBPK in cirrhosis patients was created. Simulations were performed in virtual populations and compared with clinical pharmacokinetic data.
Pharmaceutics 16 00234 g001
Figure 2. Schematic structure of the semi-PBPK model. Kti represents the gastric emptying rate and intestinal transit rate. GWi represents the gut wall of the duodenum, jejunum and ileum. kai represents the rate of drug absorption into the gut wall. QGWi represents the blood flow rate in the gut wall. QLA, QL and QPV represent the hepatic artery blood flow rate, hepatic blood flow rate and portal vein blood flow rate, respectively. CLint, CLbile and CLint,K represent the intrinsic hepatic clearance, biliary intrinsic clearance and renal intrinsic clearance, respectively.
Figure 2. Schematic structure of the semi-PBPK model. Kti represents the gastric emptying rate and intestinal transit rate. GWi represents the gut wall of the duodenum, jejunum and ileum. kai represents the rate of drug absorption into the gut wall. QGWi represents the blood flow rate in the gut wall. QLA, QL and QPV represent the hepatic artery blood flow rate, hepatic blood flow rate and portal vein blood flow rate, respectively. CLint, CLbile and CLint,K represent the intrinsic hepatic clearance, biliary intrinsic clearance and renal intrinsic clearance, respectively.
Pharmaceutics 16 00234 g002
Figure 3. The observed (points) and predicted (lines) plasma concentrations of the tested CES1 substrates and their active metabolites following intravenous or oral administration to healthy subjects. Enalapril (A) [14,104,105,106] and enalaprilat (B) [14,104,105,106] following oral 10 mg enalapril maleate; benazepril (C) [110,111] and benazeprilat (D) [110,111] following oral 20 mg benazepril hydrochloride; cilazapril (E) [51,115] and cilazaprilat (F) [51,115,116] following oral 1 mg cilazapril; perindopril (G) [119] and perindoprilat (H) [119] following oral 4 mg perindopril tert-butylamine; perindoprilat (I) [122] following oral 8 mg perindopril tert-butylamine; temocapril (J) [123] and temocaprilat (K) [123] following 1 mg temocapril hydrochloride; oseltamivir (L) [124,125,126] and oseltamivir carboxylate (M) [124,125,126] following oral 75 mg oseltamivir phosphate; flumazenil following intravenous 2.5 mg/0.5 min (N) [81] and oral 30 mg (O) [130]; pethidine following intravenous 70 mg/2 min pethidine hydrochloride (P) [134,135] and oral 100 mg pethidine hydrochloride (Q) [138]; remimazolam (R) [76] following intravenous 0.1 mg/kg remimazolam besylate. Shaded areas indicate the 5th and 95th percentiles of simulations derived from 1000 virtual individuals. The dashed lines indicate the mean of the simulated profiles.
Figure 3. The observed (points) and predicted (lines) plasma concentrations of the tested CES1 substrates and their active metabolites following intravenous or oral administration to healthy subjects. Enalapril (A) [14,104,105,106] and enalaprilat (B) [14,104,105,106] following oral 10 mg enalapril maleate; benazepril (C) [110,111] and benazeprilat (D) [110,111] following oral 20 mg benazepril hydrochloride; cilazapril (E) [51,115] and cilazaprilat (F) [51,115,116] following oral 1 mg cilazapril; perindopril (G) [119] and perindoprilat (H) [119] following oral 4 mg perindopril tert-butylamine; perindoprilat (I) [122] following oral 8 mg perindopril tert-butylamine; temocapril (J) [123] and temocaprilat (K) [123] following 1 mg temocapril hydrochloride; oseltamivir (L) [124,125,126] and oseltamivir carboxylate (M) [124,125,126] following oral 75 mg oseltamivir phosphate; flumazenil following intravenous 2.5 mg/0.5 min (N) [81] and oral 30 mg (O) [130]; pethidine following intravenous 70 mg/2 min pethidine hydrochloride (P) [134,135] and oral 100 mg pethidine hydrochloride (Q) [138]; remimazolam (R) [76] following intravenous 0.1 mg/kg remimazolam besylate. Shaded areas indicate the 5th and 95th percentiles of simulations derived from 1000 virtual individuals. The dashed lines indicate the mean of the simulated profiles.
Pharmaceutics 16 00234 g003
Figure 4. The observed (points) and predicted (lines) plasma concentrations of the tested CES1 substrates and their active metabolites following intravenous or oral administration to LC patients. Enalapril (A,C) [14,107] and enalaprilat (B,D) [14,107] following oral 10 mg enalapril maleate to CP-B (A,B) [107] and CP-C (C,D) [14]; benazepril (E) [112] and benazeprilat (F) [112] following oral 20 mg benazepril hydrochloride to CP-B; cilazapril (G) [115] and cilazaprilat (H) [115] following oral 1 mg cilazapril to CP-B; temocapril (I) [123] and temocaprilat (J) [123] following oral 1 mg temocapril hydrochloride to CP-B; oseltamivir (K) [15] and oseltamivir carboxylate (L) [15] following oral 75 mg oseltamivir phosphate to CP-B; flumazenil following intravenous 2 mg/1 min to CP-C (M) [130], 2 mg/5 min to CP-C (N) [129] and CP-B (O) [129]; flumazenil (P) [130] following oral 30 mg to CP-C; pethidine following intravenous 0.8 mg/kg,1 min (Q) [139], 0.8 mg/kg, 5 min (R) [140], 0.8 mg/kg (S,T) [136,137] pethidine hydrochloride to CP-A (Q,R,S) [136,139,140] and CP-B (T) [137]; pethidine following oral 0.8 mg/kg pethidine hydrochloride to CP-A (U) [140], 1.6 mg/kg pethidine hydrochloride to CP-A (V) [136] and CP-B (W) [137]; remimazolam following intravenous 0.1 mg/kg remimazolam besylate to CP-B (X) [141] and CP-C (Y) [141]. Shaded areas indicate the 5th and 95th percentiles of simulations derived from 1000 virtual individuals. The dashed lines indicate the mean of the simulated profiles. Comparison of the predicted AUC (Z) and Cmax (Z1) with observations in healthy subjects and LC patients. Solid, dashed and dotted lines respectively represent unity, 0.8–1.25-fold and 0.5–2-fold errors between observed and predicted data, respectively.
Figure 4. The observed (points) and predicted (lines) plasma concentrations of the tested CES1 substrates and their active metabolites following intravenous or oral administration to LC patients. Enalapril (A,C) [14,107] and enalaprilat (B,D) [14,107] following oral 10 mg enalapril maleate to CP-B (A,B) [107] and CP-C (C,D) [14]; benazepril (E) [112] and benazeprilat (F) [112] following oral 20 mg benazepril hydrochloride to CP-B; cilazapril (G) [115] and cilazaprilat (H) [115] following oral 1 mg cilazapril to CP-B; temocapril (I) [123] and temocaprilat (J) [123] following oral 1 mg temocapril hydrochloride to CP-B; oseltamivir (K) [15] and oseltamivir carboxylate (L) [15] following oral 75 mg oseltamivir phosphate to CP-B; flumazenil following intravenous 2 mg/1 min to CP-C (M) [130], 2 mg/5 min to CP-C (N) [129] and CP-B (O) [129]; flumazenil (P) [130] following oral 30 mg to CP-C; pethidine following intravenous 0.8 mg/kg,1 min (Q) [139], 0.8 mg/kg, 5 min (R) [140], 0.8 mg/kg (S,T) [136,137] pethidine hydrochloride to CP-A (Q,R,S) [136,139,140] and CP-B (T) [137]; pethidine following oral 0.8 mg/kg pethidine hydrochloride to CP-A (U) [140], 1.6 mg/kg pethidine hydrochloride to CP-A (V) [136] and CP-B (W) [137]; remimazolam following intravenous 0.1 mg/kg remimazolam besylate to CP-B (X) [141] and CP-C (Y) [141]. Shaded areas indicate the 5th and 95th percentiles of simulations derived from 1000 virtual individuals. The dashed lines indicate the mean of the simulated profiles. Comparison of the predicted AUC (Z) and Cmax (Z1) with observations in healthy subjects and LC patients. Solid, dashed and dotted lines respectively represent unity, 0.8–1.25-fold and 0.5–2-fold errors between observed and predicted data, respectively.
Pharmaceutics 16 00234 g004
Figure 5. AUCR was calculated from AUC (cirrhotic/healthy) or CL (healthy/cirrhotic) for cirrhotic status and healthy individuals, with the vast majority of parameters in the 0.5–2-fold range. (A) Enalapril; (B) enalaprilat; (C) benazepril; (D) benazeprilat; (E) I cilazapril; (F) cilazaprilat; (G) perindopril; (H) perindoprilat; (I) temocaprilat; (J) oseltamivir; (K) oseltamivir carboxylate; (L) flumazenil; (M) pethidine. Parameters not reported in the literature were excluded from the calculations; multiple doses were dose-normalized.
Figure 5. AUCR was calculated from AUC (cirrhotic/healthy) or CL (healthy/cirrhotic) for cirrhotic status and healthy individuals, with the vast majority of parameters in the 0.5–2-fold range. (A) Enalapril; (B) enalaprilat; (C) benazepril; (D) benazeprilat; (E) I cilazapril; (F) cilazaprilat; (G) perindopril; (H) perindoprilat; (I) temocaprilat; (J) oseltamivir; (K) oseltamivir carboxylate; (L) flumazenil; (M) pethidine. Parameters not reported in the literature were excluded from the calculations; multiple doses were dose-normalized.
Pharmaceutics 16 00234 g005
Figure 6. CmaxR was calculated from Cmax for cirrhotic status and healthy individuals (cirrhotic/healthy), with the vast majority of parameters in the 0.5–2-fold range. (A) Enalapril; (B) enalaprilat; (C) benazepril; (D) benazeprilaI (E) cilazapril; (F) cilazaprilat; (G) perindoprilat; (H) temocaprilat; (I) oseltamivir; (J) oseltamivir carboxylate; (K) flumazenil. Parameters not reported in the literature were excluded from the calculations; multiple doses were dose-normalized.
Figure 6. CmaxR was calculated from Cmax for cirrhotic status and healthy individuals (cirrhotic/healthy), with the vast majority of parameters in the 0.5–2-fold range. (A) Enalapril; (B) enalaprilat; (C) benazepril; (D) benazeprilaI (E) cilazapril; (F) cilazaprilat; (G) perindoprilat; (H) temocaprilat; (I) oseltamivir; (J) oseltamivir carboxylate; (K) flumazenil. Parameters not reported in the literature were excluded from the calculations; multiple doses were dose-normalized.
Pharmaceutics 16 00234 g006
Figure 7. Sensitivity analysis of enalapril and enalaprilat following oral 10 mg enalapril maleate. Enalapril: (A) Kt; (B) CLint,L; (C) GFR; (D) QLA; (E) QPV; (F) QK; (G) fu,b; (H) Peff; Enalaprilat: (I) Kt; (J) CLint,L; (K) GFR; (L) QLA; (M) QPV; (N) QK; (O) fu,b,m; (P) Peff. fu,b varies by 0.7-fold and 1.3-fold; fu,b,m varies by 0.7-fold and 1.3-fold; GFR varies by 0.5-fold and 1.5-fold; Kt, QPV and QK are varied by 1/2-fold and 2-fold; and the rest are varied by 1/3-fold and 3-fold. Individual contributions of LC-induced alterations in Kt, CES1 activity, GFR, fu,b, Peff, QK and QPV to plasma concentrations of enalapril (Q) and enalaprilat (R) following oral 10 mg enalapril maleate administration to LC patients and their integrated effects.
Figure 7. Sensitivity analysis of enalapril and enalaprilat following oral 10 mg enalapril maleate. Enalapril: (A) Kt; (B) CLint,L; (C) GFR; (D) QLA; (E) QPV; (F) QK; (G) fu,b; (H) Peff; Enalaprilat: (I) Kt; (J) CLint,L; (K) GFR; (L) QLA; (M) QPV; (N) QK; (O) fu,b,m; (P) Peff. fu,b varies by 0.7-fold and 1.3-fold; fu,b,m varies by 0.7-fold and 1.3-fold; GFR varies by 0.5-fold and 1.5-fold; Kt, QPV and QK are varied by 1/2-fold and 2-fold; and the rest are varied by 1/3-fold and 3-fold. Individual contributions of LC-induced alterations in Kt, CES1 activity, GFR, fu,b, Peff, QK and QPV to plasma concentrations of enalapril (Q) and enalaprilat (R) following oral 10 mg enalapril maleate administration to LC patients and their integrated effects.
Pharmaceutics 16 00234 g007
Table 1. Physiological parameters used in the physiologically based pharmacokinetic model in adults with and without cirrhosis.
Table 1. Physiological parameters used in the physiologically based pharmacokinetic model in adults with and without cirrhosis.
NormalChild–Pugh ClassUnits
ABC
Blood flow rates
  Liver a1450 [17,18]1436.51176.91656.3mL/min
  Hepatic arterial300 [18]390 [17,18]486.9 [9]1020 [17]mL/min
  Portal vein1150 [18]1046.5 [9]690 [19]636.3 [9]mL/min
  Kidney1240 [18]1091.2 [17]806 [17]595.2 [17]mL/min
  Duodenum b45 [20]454545mL/min
  Jejunum b173 [20]173173173mL/min
  Ileum b102 [20]102102102mL/min
Volume
  Liver1690 [18]1368.9 [21]1098.5 [21]895.7 [21]mL
  Portal vein b70 [18]707070mL
  Kidney b280 [18]280280280mL
  Duodenum b21 [22]212121mL
  Jejunum b63 [22]636363mL
  Ileum b42 [22]424242mL
  Transit rates c
Stomach0.04 [23]0.0504 [24]0.0504 [24]0.0504 [24]min−1
  Duodenum0.07 [23]0.0889 [24]0.0889 [24]0.0889 [24]min−1
  Jejunum0.03 [23]0.0381 [24]0.0381 [24]0.0381 [24]min−1
  Ileum0.04 [23]0.0508 [24]0.0508 [24]0.0508 [24]min−1
Gut radius
  r1 b2 [23]222cm
  r2 b1.63 [23]1.631.631.63cm
  r3 b1.45 [23]1.451.451.45cm
Glomerular filtration rate105 [25]82 [25]82 [25]82 [25]mL/min
Albumin44.7 [9]36.2 [17]30.4 [17]26.3 [9]g/L
α1-acid glycoprotein0.8 [21]0.57 [21]0.52 [21]0.46 [21]g/L
CES12.45 [9]2.45 [9]1.715 [9]0.735 [9]mg/g Liver
CYP2B617 [21]17 [21]15.3 [21]13.6 [21]pmol/mg
Lactulose/Rhamnose ratio0.037 [26]0.046 [26]0.052 [26]0.057 [26]/
MRP2 ratio10.54 [19]0.54 [19]0.54 [19]/
a: QL = QLA + QPV, hepatic blood flow rate equals hepatic arterial blood flow rate plus portal vein blood flow rate. b: Assuming that the values are unchanged in cirrhosis. c: Transit rates in cirrhosis were corrected by Table 1 of reference [24].
Table 2. Simultaneously predicting the pharmacokinetics of CES1-metabolized drugs and their metabolites using the physiologically based pharmacokinetic model.
Table 2. Simultaneously predicting the pharmacokinetics of CES1-metabolized drugs and their metabolites using the physiologically based pharmacokinetic model.
DruglogPpkaCLintVmaxKmKL;P dKG;P dKK;P dCLbVsysK12K21Peff,A–BCLint,KRbfu,bFka
mL/minnmoL/min/
mg protein
μmol/L mL/minLmin−1min−110−4 cm/smL/min
Enalapril0.59 [27]5.20 [27]784 [28]//1.662.291.79/40 [29]//1.60 [30]624.6 [31]0.74 [32]0.74 [33]/
Enalaprilat−0.74 [33]2.03 [33]///1.121.041.25/46.1 [34]0.001 [34]0.0009 [34]/186.4 [35]0.73 [32]0.68 [33]/
Oseltamivir0.36 [36]7.7 [36]20,255.4 [36]//1.191.121.29/61.289 [37] f///1357.95 [38]1 e0.58 [36]/0.061 [39] g
Oseltamivir
carboxylate
−1.3 a4.19 a///1.711.891.91/160.729 [40] f///438.5 [41]1 e0.97 [36]/
Benazepril1.11 [42]4.74 [42]6696 [43]//0.0870.1220.088385.8 [44] g4.8 [45] g0.0215 [45] g0.0238 [45] g1.21 [46]8391.6 c1 e0.03 [47]0.35 [29]
Benazeprilat0.56 [42]1.97 [42]///0.0930.0880.101/1.204 [48] f0.0438 [48] f0.00837 [48] f/447.9 [47]1 e0.05 [47]/
Cilazapril0.55 [49]3.3 [50]199.7 c//1.321.311.43205 a18.23 [51] f0.00325 [51] f0.00155 [51] f/118.095 [52]1 e0.7 [49]/0.099 [53] g
Cilazaprilat−0.48 a3.17 a///1.281.221.42/10.3517 [51] f0.00084 [51] f0.008 [51] f/75.48 [52]1 e0.76 [54]//
Temocapril2.102 [55]2.8 [56]5359.7 [57]//2.823.172.47/15.398 [58] g///110.2 [59]1 e0.3 [60]0.65 [61]0.065 [58] g
Temocaprilat2.215 [62]2.09 [60]///0.2890.3220.251/58.535 [63] f0.00184 [63] f0.000078 [63] f/949.84 [64]
1899.68 [65] b
1 e0.025 [60]//
Perindopril−1.31 [66]3.2 [67]1011.15 [68]
156.47 [69] c,j
//0.6650.6330.742/13.119 [70] g0.0028 [70] g0.0024 [70] g1.34 [43]130.2 [71]1 e0.4 [72]0.66 [64]/
Perindoprilat−0.08 a3.08 a///1.451.381.61/53.44 [73] f0.271 [73] f0.0996 [73] f/231.78 [74]1 e0.85 [72]//
Remimazolam3.68 a5.99 a79,212.96 c//36.3463.1931.21180 [75]15.0768 [76] f0.01638 [76] f
0.3117 [76] f (K13)
0.000476 [76] f
0.5057 [76] f (K31)
//1 e0.08 [77]/
Flumazenil1.64 [78]0.86 [79]8169.9 c//2.572.712.411120 [80]24.054 [81] g0.0376 [81] g0.0427 [81] g3.78 [82]1.67 [83]1 [84]0.6 [85]/
Pethidine2.35 [86]8.7 [86]/1.56 [87] h
5.382 [88] i
261 [87] h
356 [88] i
14.824.1812.02/328.676 [89] f0.002224 [89] f0.0003697 [89] f/58.78 [90]0.87 [91]0.48 [88]/0.117 [92] g
a: Data from www.drugbank.com, accessed on 4 February 2024; b: Bile intrinsic clearance of temocaprilat; c: Recalculated from CLL,b; d: Calculations using Rodgers–Rowland method; e: Assumed values; f: Simulation by WinNonlin, cilazapril and cilazaprilat using 0.5 mg dose pharmacokinetic and remimazolam using 0.025 mg/kg dose pharmacokinetic in simulation; g: Calculated by WinNonlin, flumazenil using T.F. pharmacokinetic to calculate; h: CES1-mediated CLint; i: CYP2B6-mediated CLint; j: UGT intrinsic clearance of perindopril.
Table 4. Observed and predicted values of AUC0–t and Cmax of enalapril and enalaprilat following oral enalapril maleate administration to healthy (HT) subjects and liver cirrhosis patients.
Table 4. Observed and predicted values of AUC0–t and Cmax of enalapril and enalaprilat following oral enalapril maleate administration to healthy (HT) subjects and liver cirrhosis patients.
DrugDoseSubjectsAUC0–t (μg × h/mL)Cmax (ng/mL)
ObsPreObs/PreObsPreObs/Pre
Enalapril10 mg [14]HT0.12290.14670.8466.945.61.47
10 mg [104]HTNR0.1151/NR45.6/
10 mg [105]HT0.16000.15261.0572.145.61.58
10 mg [105]HT0.14800.15470.9665.445.61.43
10 mg [106]HTNR0.1467/NR45.6/
10 mg [107]CP-B0.17610.22530.78110.160.61.82
10 mg [14]CP-C0.27690.31950.87123.480.71.53
Enalaprilat10 mg [14]HT0.37540.36831.0246.139.71.16
10 mg [104]HTNR0.3683/NR39.7/
10 mg [105]HT0.21700.36830.5929.339.70.74
10 mg [105]HT0.26000.36830.7137.339.70.94
10 mg [106]HTNR0.2776/NR39.7/
10 mg [107]CP-B0.38120.51540.7436.835.11.05
10 mg [14]CP-C0.17330.24760.7016.820.10.84
NR: Not reported.
Table 5. Observed and predicted values of AUC0–t and Cmax of benazepril and benazepril following benazepril hydrochloric administration to healthy (HT) subjects and cirrhosis.
Table 5. Observed and predicted values of AUC0–t and Cmax of benazepril and benazepril following benazepril hydrochloric administration to healthy (HT) subjects and cirrhosis.
DrugDoseSubjectsAUC0–t (μg × h/mL)Cmax (ng/mL)
ObsPreObs/PreObsPreObs/Pre
Benazepril10 mg [108]HT0.13900.25710.54139.139113.5451.23
10 mg [109]HT0.13800.26650.5278.957113.5450.70
20 mg [110]HT0.21950.46110.48265.313227.0891.17
20 mg [111]HTNR0.4611/252.98227.0891.11
20 mg [112]CP-B0.61590.58831.05543.472268.1302.03
Benazeprilat10 mg [113]HT1.53301.64920.93188.704198.970.95
10 mg [108]HT1.07871.35540.80200.410198.971.01
10 mg [109]HT1.10391.35540.81164.520198.970.83
20 mg [110]HT2.38002.71070.88463.830397.951.17
20 mg [111]HTNR2.7107/342.164397.950.86
20 mg [112]CP-B2.16502.38700.91345.010344.851.00
NR: Not reported.
Table 6. Observed and predicted values of AUC0–t and Cmax of cilazapril following oral cilazapril to healthy (HT) subjects and LC patients.
Table 6. Observed and predicted values of AUC0–t and Cmax of cilazapril following oral cilazapril to healthy (HT) subjects and LC patients.
DrugDoseSubjectsAUC0–t (μg × h/mL)Cmax (ng/mL)
ObsPreObs/PreObsPreObs/Pre
Cilazapril1 mg [51]HT0.09980.10440.9633.926.21.29
2.5 mg [51]HT0.25600.26100.9882.765.41.26
5 mg [51]HT0.49600.52210.95182.0130.81.39
2.5 mg [114]HT0.18300.23410.7875.765.41.16
1 mg [115]HT0.06570.08900.7425.226.20.96
1 mg [115]CP-B0.18400.12011.5340.028.31.41
Cilazaprilat1 mg [51]HT0.07910.07251.0912.410.21.22
1 mg [116]HTNR0.1158/8.310.20.81
2.5 mg [51]HT0.1750.18110.9737.725.41.48
5 mg [51]HT0.3420.36230.9494.250.81.85
2.5 mg [114]HT0.1780.18110.9839.325.41.55
5 mg [117]HT0.3980.65800.6083.450.81.64
1.25 mg [118]HT0.0700.09060.7713.012.71.02
2.5 mg [118]HT0.1700.18110.9436.025.41.42
5 mg [118]HT0.2800.36230.7774.050.81.46
10 mg [118]HT0.5500.72460.76165.0101.51.63
1 mg [115]HT0.0530.07250.737.9610.20.78
1 mg [115]CP-B0.07750.06951.1210.28.31.23
NR: Not reported.
Table 7. Observed and predicted values of AUC0t and Cmax of perindopril following oral perindopril tert-butylamine administration to healthy (HT) subjects and LC patients.
Table 7. Observed and predicted values of AUC0t and Cmax of perindopril following oral perindopril tert-butylamine administration to healthy (HT) subjects and LC patients.
DrugDoseSubjectsAUC0–t (μg × h/mL)Cmax (ng/mL)
ObsPreObs/PreObsPreObs/Pre
Perindopril4 mg [119]HT0.1210.1201.0164.234.61.86
8 mg [120]CP-A0.3770.2391.58NR70.4/
8 mg [121]CP-B0.6020.2812.14NR77.0/
Perindoprilat4 mg [119]HT0.05200.06810.764.74.31.09
8 mg [122]HT0.11970.13620.88NR8.5/
8 mg [120]CP-A0.32100.26951.19298.83.33
8 mg [121]CP-B0.13400.27770.48NR8.6/
NR: Not reported.
Table 8. Observed and predicted values of AUC0–t and Cmax of temocapril and temocaprilat following oral temocapril hydrochloride administration to healthy (HT) subjects and LC patients.
Table 8. Observed and predicted values of AUC0–t and Cmax of temocapril and temocaprilat following oral temocapril hydrochloride administration to healthy (HT) subjects and LC patients.
DrugDoseSubjectsAUC0–t (μg × h/mL)Cmax (ng/mL)
ObsPreObs/OreObsPreObs/Ore
Temocapril1 mg [123]HTNR0.0257/NR11.0/
1 mg [123]CP-BNR0.0271/NR11.6/
Temocaprilat1 mg [123]HT0.12300.11991.0315.811.21.41
1 mg [123]CP-B0.17140.08002.1414.37.41.93
NR: Not reported.
Table 9. Observed and predicted values of AUC0t and Cmax of oseltamivir and oseltamivir carboxylate (OC) following oral oseltamivir phosphate administration to healthy (HT) subjects and cirrhosis.
Table 9. Observed and predicted values of AUC0t and Cmax of oseltamivir and oseltamivir carboxylate (OC) following oral oseltamivir phosphate administration to healthy (HT) subjects and cirrhosis.
DrugDoseSubjectsAUC0–t (μg × h/mL)Cmax (ng/mL)
ObsPreObs/PreObsPreObs/Pre
Oseltamivir75 mg [126]HT0.15900.14301.1174.459.81.24
75 mg [125]HT0.12400.14300.8775.159.81.26
75 mg [125]HT0.11400.14300.8067.659.81.13
75 mg [124]HT0.11880.14420.8261.059.81.02
150 mg [126]HT0.31300.28601.09192.0119.51.61
75 mg [15]CP-B0.21000.19851.06100.085.61.17
Oseltamivir carboxylate75 mg [126]HT3.02002.50681.20291.00264.931.10
75 mg [125]HT2.65002.50681.06276.00264.931.04
75 mg [125]HT2.56002.50681.02278.00264.931.05
75 mg [124]HT3.17633.08611.03360.31264.931.36
150 mg [126]HT6.31005.01351.26550.00529.861.04
75 mg [15]CP-B3.10004.32350.72260.00279.860.93
Table 10. Observed and predicted values of AUC0–t (μg × h/mL) or CL (L/min) and Cmax (ng/mL) of flumazenil to healthy (HT) subjects and LC patients.
Table 10. Observed and predicted values of AUC0–t (μg × h/mL) or CL (L/min) and Cmax (ng/mL) of flumazenil to healthy (HT) subjects and LC patients.
DoseSubjectsAUC0–t or CLCmax
ObsPreObs/PreObsPreObs/Pre
10 mg, 1 min, i.v. [127]HT0.9000 a0.4486 a2.01
10 mg, 10 min, i.v. [128]HT0.8967 a0.4549 a1.97
2.5 mg, 0.5 min, i.v. [81]HT0.7160 a0.4766 a1.50
2 mg, 5 min, i.v. [129]CP-B0.4932 a0.4988 a0.99
2 mg, 5 min, i.v. [129]CP-C0.3165 a0.4030 a0.79
2 mg, 1 min, i.v. [130]CP-C0.7050 a0.4295 a1.64
30 mg, p.o. [130]HTNR0.1741 b/70.171.00.99
30 mg, p.o. [130]CP-CNR0.5139 b/258.0174.71.48
a: represent CL; b: represent AUC; NR: Not reported.
Table 11. Observed and predicted values of AUC0–t (μg × h/mL) or CL (L/min) and Cmax (ng/mL) of pethidine following oral and intravenous pethidine HCl administration to healthy (HT) subjects and LC patients.
Table 11. Observed and predicted values of AUC0–t (μg × h/mL) or CL (L/min) and Cmax (ng/mL) of pethidine following oral and intravenous pethidine HCl administration to healthy (HT) subjects and LC patients.
DoseSubjectsAUC0–t or CLCmax
ObsPreObs/PreObsPreObs/Pre
25 mg, 1 min, i.v. [131]HT0.5624 a0.4956 a1.13
50 mg, 1 min, i.v. [132]HT1.0200 a0.7784 a1.31
50 mg, 1 min, i.v. [133]HT0.9640 a0.8532 a1.13
70 mg, 2 min, i.v. [134]HT0.7505 a0.4952 a1.52
70 mg, 2 min, i.v. [135]HT0.7226 a0.4952 a1.46
0.8 mg/kg, 1 min, i.v. [139]HT1.3160 a0.7887 a1.67
0.8 mg/kg, 5 min, i.v. [140]HT0.9000 a0.6972 a1.29
0.8 mg/kg, 1 min, i.v. [136]CP-A0.3920 a0.5349 a0.73
0.8 mg/kg, 1 min, i.v. [139]CP-A0.6640 a0.7405 a0.90
0.8 mg/kg, 5 min, i.v. [140]CP-A0.5730 a0.7560 a0.76
0.8 mg/kg, 1 min, i.v. [137]CP-B0.3730 a0.5724 a0.65
25 mg, p.o. [131]HT0.9270 a0.8563 a1.08NR36.0/
100 mg, p.o. [138]HT0.8600 b0.6097 b1.41170.0143.91.18
0.8 mg/kg, p.o. [140]HTNR0.4649 b/NR80.6/
1.6 mg/kg, p.o. [136]CP-ANR1.2681 b/NR157.0/
0.8 mg/kg, p.o. [140]CP-ANR0.4439 b/NR78.5/
1.6 mg/kg, p.o. [137]CP-BNR1.1983 b/NR146.3/
a: represent CL; b: represent AUC; NR: Not reported.
Table 12. Observed and predicted values of AUC0–t of remimazolam following intravenous remimazolam besylate administration to healthy (HT) subjects and LC patients.
Table 12. Observed and predicted values of AUC0–t of remimazolam following intravenous remimazolam besylate administration to healthy (HT) subjects and LC patients.
DoseSubjectsAUC0–t (μg × h/mL)
ObsPreObs/Pre
0.05 mg/kg [76]HT0.04470.05360.83
0.075 mg/kgHT0.06650.07870.84
0.1 mg/kgHT0.08600.10000.86
0.2 mg/kgHT0.16830.21450.78
0.3 mg/kgHT0.25170.29600.85
0.4 mg/kgHT0.33170.39790.83
10.4 mg [141]CP-BNR0.1277/
8.2 mgCP-CNR0.0805/
NR: Not reported.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Luo, X.; Zhang, Z.; Mu, R.; Hu, G.; Liu, L.; Liu, X. Simultaneously Predicting the Pharmacokinetics of CES1-Metabolized Drugs and Their Metabolites Using Physiologically Based Pharmacokinetic Model in Cirrhosis Subjects. Pharmaceutics 2024, 16, 234. https://doi.org/10.3390/pharmaceutics16020234

AMA Style

Luo X, Zhang Z, Mu R, Hu G, Liu L, Liu X. Simultaneously Predicting the Pharmacokinetics of CES1-Metabolized Drugs and Their Metabolites Using Physiologically Based Pharmacokinetic Model in Cirrhosis Subjects. Pharmaceutics. 2024; 16(2):234. https://doi.org/10.3390/pharmaceutics16020234

Chicago/Turabian Style

Luo, Xin, Zexin Zhang, Ruijing Mu, Guangyu Hu, Li Liu, and Xiaodong Liu. 2024. "Simultaneously Predicting the Pharmacokinetics of CES1-Metabolized Drugs and Their Metabolites Using Physiologically Based Pharmacokinetic Model in Cirrhosis Subjects" Pharmaceutics 16, no. 2: 234. https://doi.org/10.3390/pharmaceutics16020234

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop