kobe university repository : kernel7 1department of clinical pharmacy and education, graduate school...

39
Kobe University Repository : Kernel タイトル Title Population pharmacokinetics and pharmacodynamics of mycophenolic acid using the prospective data in patients undergoing hematopoietic stem cell transplantation 著者 Author(s) Yoshimura, Kazuaki / Yano, Ikuko / Yamamoto, Takashi / Kawanishi, Misaki / Isomoto, Yui / Yonezawa, Atsushi / Kondo, Tadakazu / Takaori- Kondo, Akifumi / Matsubara, Kazuo 掲載誌・巻号・ページ Citation Bone Marrow Transplantation,53(1):44-51 刊行日 Issue date 2018-01 資源タイプ Resource Type Journal Article / 学術雑誌論文 版区分 Resource Version author 権利 Rights © 2017, Springer Nature DOI 10.1038/bmt.2017.213 JaLCDOI URL http://www.lib.kobe-u.ac.jp/handle_kernel/90004835 PDF issue: 2020-11-06

Upload: others

Post on 10-Aug-2020

2 views

Category:

Documents


0 download

TRANSCRIPT

Kobe University Repository : Kernel

タイトルTit le

Populat ion pharmacokinet ics and pharmacodynamics of mycophenolicacid using the prospect ive data in pat ients undergoing hematopoiet icstem cell t ransplantat ion

著者Author(s)

Yoshimura, Kazuaki / Yano, Ikuko / Yamamoto, Takashi / Kawanishi,Misaki / Isomoto, Yui / Yonezawa, Atsushi / Kondo, Tadakazu / Takaori-Kondo, Akifumi / Matsubara, Kazuo

掲載誌・巻号・ページCitat ion Bone Marrow Transplantat ion,53(1):44-51

刊行日Issue date 2018-01

資源タイプResource Type Journal Art icle / 学術雑誌論文

版区分Resource Version author

権利Rights © 2017, Springer Nature

DOI 10.1038/bmt.2017.213

JaLCDOI

URL http://www.lib.kobe-u.ac.jp/handle_kernel/90004835

PDF issue: 2020-11-06

1

Population pharmacokinetics and pharmacodynamics of mycophenolic acid using prospective data in 1

patients undergoing hematopoietic stem cell transplantation 2

3

Kazuaki Yoshimura1,2, Ikuko Yano1,2,3, Takashi Yamamoto2, Misaki Kawanishi1,2, Yui Isomoto2, Atsushi 4

Yonezawa1,2, Tadakazu Kondo4, Akifumi Takaori-Kondo4, and Kazuo Matsubara2 5

6

1Department of Clinical Pharmacy and Education, Graduate School of Pharmaceutical Science, Kyoto 7

University, Sakyo-ku, Kyoto 606-8501, Japan. 8

2Department of Clinical Pharmacology and Therapeutics, Kyoto University Hospital, Sakyo-ku, Kyoto 9

606-8507, Japan. 10

3Department of Pharmacy, Kobe University Hospital, Chuo-ku, Kobe 650-0017, Japan. 11

4Department of Hematology and Oncology, Graduate School of Medicine, Kyoto University, Sakyo-ku, 12

Kyoto 606-8507, Japan 13

14

Running heading: 15

Population analysis of mycophenolic acid 16

2

17

Conflict of interest: The authors declare no conflict of interest. 18

19

This study was supported in part by JSPS KAKENHI Grant Number JP25460210. 20

21

Corresponding Author: Ikuko Yano, PhD 22

Department of Pharmacy, Kobe University Hospital, 23

Chuo-ku, Kobe 650-0017, Japan 24

Tel.: +81-78-382-6641 25

Fax: +81-78-382-6676 26

E-mail Address: [email protected] 27

28

29

30

3

Abstract 31

Mycophenolate mofetil (MMF), a prodrug of mycophenolic acid (MPA), is used to suppress 32

graft-versus-host disease in patients undergoing hematopoietic stem cell transplantation (HCT). The 33

purpose of this study was to construct a population pharmacokinetic and pharmacodynamic model in 34

HCT patients for individualized MPA therapy. Blood samples were obtained from 49 HCT patients after 35

starting MMF therapy. Population pharmacokinetic and pharmacodynamic parameters were obtained 36

using the program NONMEM. MPA was described via a 1-compartment model with a first order 37

elimination, and 30.9% of MPA glucuronide (MPAG) was found in the enterohepatic circulation. 38

Inosine-5’-monophosphate dehydrogenase (IMPDH) activity was modeled as a maximal inhibitory model 39

with a half-maximal inhibitory concentration (IC50) of 3.59 µg/mL against MPA concentrations. 40

Simulations based on the obtained pharmacokinetic and pharmacodynamic parameters revealed that 41

decreased creatinine clearance increases the MPAG concentration followed by an increased MPA 42

concentration; therefore, IMPDH activity decreases. Diarrhea decreases the enterohepatic circulation of 43

MPAG and consequently reduces MPA concentration. The IC50 for MPA exhibited a positive association 44

with C-reactive protein. Dosage adjustment based on plasma MPA concentration is required especially 45

for patients with renal dysfunction and/or diarrhea. 46

4

Introduction 47

Mycophenolate mofetil (MMF) is clinically used to suppress graft-versus-host disease (GVHD) in 48

patients undergoing hematopoietic stem cell transplantation (HCT) and acute rejection after solid organ 49

transplantation1,2. Mycophenolic acid (MPA), an active form of MMF, is metabolized by 50

glucuronosyltransferases in the liver. MPA glucuronide (MPAG) and MPA acyl glucuronide (AcMPAG) 51

are primarily produced by UGT1A9 and 2B7, respectively 3. While MPAG is an inactive metabolite, 52

AcMPAG exhibits pharmacological activity in vitro and is potentially responsible for the toxicity of MPA 53

4. The majority of MPA metabolites are eliminated via the urine and partial elimination also occurs in the 54

bile mediated by multidrug resistance associated protein 2 (MRP2) followed by the entero-hepatic 55

recirculation 5. 56

The pharmacokinetics (PK) of MPA exhibits a large inter- and intra-individual variability, and it is 57

recommended that the area under the concentration-time curve (AUC) of MPA be monitored for 58

individualized therapy in solid organ transplant recipients6, 7. Recently, Arai et al. 8 proposed that MPA 59

drug monitoring was necessary for the effective prophylaxis of acute GVHD undergoing cord blood stem 60

cell transplantation (CBT). However, information regarding the optimal dose of MMF or the target range 61

for MPA concentrations in HCT patients is limited 9. 62

5

MPA selectively inhibits inosine-5’-monophosphate dehydrogenase (IMPDH) and suppresses the 63

proliferation of B and T lymphocytes 10. IMPDH exists as two isoforms derived from different genes 11, 12, 64

and recombinant proteins of IMPDH2 is 4.8-fold more sensitive to MPA than IMPDH113. The area under 65

the effect curve (AUEC) of IMPDH activity on day 21 after HCT was reportedly associated with both 66

non-relapse and overall mortality 14. Therefore, the measurement of IMPDH activity in peripheral blood 67

mononuclear cells (PBMCs) in addition to monitoring the AUC of MPA is considered to be an effective 68

predictor of the clinical outcome of MPA therapy. 69

The PK of MPA is influenced by serum albumin, renal dysfunction, total bilirubin, age, 70

co-administration with cyclosporine, and dose 15-19. In addition, the incidence of acute rejection in the first 71

year post-transplantation was significantly lower in carriers of SNPs for IMPDH1 -106 G>A and 125 72

G>A compared with the respective wild-type individuals20. The SNP for IMPDH2 3757 T>C was 73

associated with a significantly higher IMPDH activity following the MMF intake, despite of no difference 74

in the MPA exposure between groups21. The SNP for MRP2 -24C>T was associated with a significantly 75

higher dose-corrected MPA trough levels at later time points after transplantation22. The SNP for 76

UGT2B7 -842G>A resulted in a significantly higher AUC of AcMPAG at 1 and 3 months 77

post-transplantation in patients with renal transplantation23. 78

6

In this study, the effects of the patient characteristics including previously proven genetic 79

polymorphisms were examined using a population PK and pharmacodynamics (PD) analysis. Effects of 80

covariates were quantitatively evaluated by the simulation to examine the clinical significance of these 81

covariates. 82

83

Subjects and Methods 84

Study design 85

A total of 49 adult Japanese HCT patients between March 2013 and August 2016 were included in 86

the study. Acute GVHD prophylaxis comprised tacrolimus (PrografTM, Astellas Pharma Inc., Tokyo, 87

Japan) and MMF (CellceptTM, Chugai Pharmaceutical Co., Ltd., Tokyo, Japan) in CBT, plus short-term 88

methotrexate (MethotrexateTM, Pfizer Japan Inc., Tokyo, Japan) in bone marrow transplant (BMT) or 89

peripheral blood stem cell transplantation (PBT). MMF was orally administered at 10 mg/kg every 8 h 90

(30 mg/kg/day), and was initiated on day −1 after CBT or on day 7 after BMT and PBT, except in one 91

patient administered 15 mg/kg every 12 h. No potentially interacting drugs including cyclosporine or 92

foods with MPA were co-administered. 93

Pre-transplant recipient DNA was used to determine UGT2B7 −842C>T (rs7439366) and MRP2 94

−24C>T (rs717620) genotypes. Approximately five weeks after MMF administration commenced, 95

7

post-transplant donor DNA as well as pre-transplant recipient DNA were used to determine IMPDH1 −96

106G>A (rs2278294), IMPDH1 125G>A (rs2278293), and IMPDH2 3757T>C (rs11706052) genotypes. 97

Blood samples were collected immediately before, 1, 2, 4, and 8 h after the first and third weeks after 98

MMF administration commenced, plus blood sampling at 12 h after MMF administration in one patient 99

administered 15 mg/kg every 12 h. This clinical study was approved by the Ethics Committee of Kyoto 100

University Graduate School and Faculty of Medicine and Kyoto University Hospital. Written informed 101

consent was obtained from all patients included in the study. 102

103

Analytical methods 104

Total plasma concentrations of MPA, MPAG, and AcMPAG were analyzed using LC-MS/MS 105

according to the previously reported method 24. The lower limits of quantification (LLOQ) were 0.05, 0.2, 106

and 0.02 µg/mL for MPA, MPAG, and AcMPAG, respectively. PBMC samples were used to measure the 107

IMPDH activity according to the previous method 24. The IMPDH activity was calculated based on the 108

XMP produced, which was normalized to the intracellular AMP. The LLOQ were 50 nM for both XMP 109

and AMP. The data for AMP under the LLOQ were excluded from the analysis due to extremely low 110

white blood cell counts after the transplantation. 111

112

Population PK/PD analysis 113

8

A population PK analysis was conducted using NONMEM. The overview of the basic PK/PD 114

model for MPA is shown in Fig. 1. Since only the oral data were available, the relative bioavailability (F) 115

of MPA was assumed to be 1. The model was parameterized using clearances for MPA, MPAG, and 116

AcMPAG (CLMPA, CLMPAG, and CLAcMPAG), as well as the volume of distribution for MPA, MPAG, and 117

AcMPAG (VMPA, VMPAG, and VAcMPAG). It was assumed that MPA was metabolized to MPAG and 118

AcMPAG by a first-order process, in which 99 % and 1 % of MPA was converted to MPAG and 119

AcMPAG, respectively, because the ratio of AUC0–8 for MPAG to AcMPAG was approximately 99:1 in 120

this study. The enterohepatic circulation (EHC) was tested as a first-order process (KEHC) from the central 121

compartment of each metabolite to the gastrointestinal tract. For the comparison, 2-compartment model 122

with EHC, and the lag time and the transit compartment models in the absorption process were tested 25. 123

Additionally, EHC modeling by presuming a hypothetical gall bladder compartment was tested 26, 27. 124

Interindividual and interoccasion variability (IIV and IOV) in the PK/PD parameters were 125

modeled using an exponential error model 28. The estimation for the IOV was as follows: occasions 1 and 126

2 pertained to one and three weeks after MMF administration commenced, respectively. The influence of 127

each covariate on the population mean parameters was evaluated by the stepwise forward inclusion and 128

backward elimination method, and significance levels were 1 % and 0.1 % (6.63 and 10.8 with freedom 129

9

of 1 assuming a chi-square distribution), respectively. The tested covariates for the PK parameters 130

included body weight, gender, stem cell source, age, aspartate aminotransferase, serum albumin, total 131

bilirubin, creatinine clearance (CLCR), dose of MMF, diarrhea, and investigated genotypes for MRP2 and 132

UGT2B7 of recipient. Diarrhea was defined as the occurrence of loose, muddy or watery stool, or more 133

than five times per a day of fecal frequency in case of not recording the fecal condition. 134

Continuous variables were normalized by each population median using the following power 135

function model: 136

θ θ 137

(1) 138

where θi is the individual model-predicted PK parameter (e.g. CLMPA) for an individual with covariate 139

value of COVi. θpop represents the population mean for the parameter θ, COVmed represents the population 140

median of the covariate, and θcov represents the covariate effect. For dichotomous variables, the value of 141

COVi is typically set to 0 for the normal classification and 1 for the other classifications in each 142

individual as follows: 143

θ θ θ (2) 144

----

10

After the final population PK model was obtained, the relationship between the MPA 145

concentrations and IMPDH activity was explored graphically and modeled using a direct sigmoid 146

inhibitory maximum effect model as followed: 147

E E 1,

(3) 148

where E0, IC50,MPA, and represent baseline of IMPDH activity, half-maximal inhibitory MPA 149

concentration, and the Hill coefficient to be estimated 29, and CMPA represents the MPA concentration. To 150

investigate the effect of the AcMPAG concentration on IMPDH activity, an additional inhibitory effect 151

model was tested 30. The tested covariates for the PD parameters included the stem cell source, 152

reduced-intensity conditioning, gender, age, serum albumin, C-reactive protein (CRP), and investigated 153

genotypes (IMPDH1 and 2) of donor or recipient. In the value of CRP was under the LLOQ (<0.2 154

mg/mL), this value was converted to 0.1 due to the difficulty of the calculation. Goodness-of-fit and 155

prediction-corrected visual predictive check plots were used for internal validation 31. For 156

prediction-corrected visual predictive check plots, the final PK/PD model was used to simulate original 157

data sets at 1000 times compared with the observed data. 158

159

Simulation study 160

11

The effects of statistically significant covariates on the PK/PD of MPA were evaluated by the 161

simulation using the final population parameters. The dose was fixed to 500 mg every 8 h for all 162

simulations. In the simulation for the effect of each covariate, other covariates were fixed to the median 163

value of each covariate and without diarrhea. The AUC0–8 or AUEC0–8 were calculated using the linear 164

trapezoidal method. 165

166

Results 167

Patient characteristics 168

The patient characteristics and the distribution of each genotype before and five weeks after the 169

transplantation are summarized in Table 1. All of the observed genotype distributions were consistent 170

with Hardy-Weinberg equilibrium. 171

172

Population PK modeling 173

In total, 522 concentration data for MPA, MPAG, and AcMPAG, respectively, were analyzed. Five 174

samples with MPA concentrations under the LLOQ were replaced with half of the LLOQ 32, and included 175

in the analysis. 2-compartment model improved the model fitting compared with 1-compartment model 176

12

without EHC. However, after an inclusion of EHC process, 2-compartment model did not improve the 177

model fitting compared with 1-compartment model (∆OBJ = −8.12), and a terminal elimination rate 178

constant was not correctly estimated. Therefore, the PK of MPA was finally described by 1-compartment 179

model with first order absorption and elimination, and was affected by the EHC of MPAG. An inclusion 180

of the absorption lag-time did not improve the model fitting. Transit compartment model was not adopted 181

owing to the high computational intensity required, although the model fit was significantly improved. 182

Although the inclusion of EHC of AcMPAG in the model brought a statistically significant model 183

improvement, CLAcMPAG was not correctly estimated. Therefore, the model including the EHC of only 184

MPAG was selected. EHC modeling by presuming a hypothetical gall bladder compartment did not 185

improve the model fitting compared with first-order EHC model. The simultaneous inclusion of IOVs for 186

Ka, F, KEHC, CLMPAG, and CLAcMPAG significantly improved the model fitting (∆OBJ = −721). The final 187

PK parameters and its relative standard error (RSE) are presented in Table 2. Figure 2 shows the 188

inter-occasional parameters for one and three weeks after MMF administration commenced. 189

After the evaluation of each covariate, the serum albumin revealed a significant negative association 190

with CLMPA and VMPA. CLCR exhibited a significant positive relationship with both CLMPAG and CLAcMPAG, 191

and KEHC in the patients showing diarrhea was 0.375-fold lower than that without diarrhea (Table 2). An 192

13

inclusion of IIV was tested for all the MPA PK parameters and an exclusion of IIV was tested after 193

inclusion of IOV. After all, IIVs on VMPA and F were retained in the final model, and shrinkage values of 194

them were 27.6% and 16.8%, respectively. The ratio for the EHC of MPAG was estimated to be 30.9% 195

(EHC (%) = KEHC/(KEHC + CLMPAG/VMPAG) × 100) in patients with CLCR of 112 mL/min without diarrhea. 196

197

PD modeling 198

A total of 460 IMPDH activity data from 49 patients were used for the PD model building following 199

the PK modeling process. The 62 IMPDH activity data were excluded due to AMP under the LLOQ. The 200

IMPDH activity was described with the inhibitory Emax model using the MPA concentrations. The Hill 201

coefficient was fixed to 1 by the statistical selection (∆OBJ = −3.00). The additive inhibitory effect 202

model for AcMPAG did not significantly improve the model fitting. An inclusion of IOV on E0 203

significantly improved the model fitting (∆OBJ = −200). The value of IC50 for MPA revealed a positive 204

association with CRP (∆OBJ = −11.4). No polymorphisms were identified as significant covariates in 205

the PK/PD model. The final PD parameters with RSE are shown in Table 2. The IIV for IC50 was 81.2%, 206

and its shrinkage was 29.7%. The goodness-of-fit and prediction-corrected visual predictive check 207

14

demonstrated that the population PK/PD model accurately predicted the observed MPA and its 208

metabolites concentrations, as well as IMPDH activity (Figs. 3 and 4). 209

210

Simulation study 211

A total of 1,000 data sets in each group were simulated under the several renal functions with or 212

without diarrhea (Fig. 5). The AUC0-8 of MPAG and AcMPAG significantly increased according to the 213

decreased CLCR compared with those for 120 mL/min. The AUC0-8 of MPA also significantly increased 214

according to the decreased CLCR. The AUEC0-8 of IMPDH significantly decreased from 339 to 215 215

µmol·h·sec− 1·mol AMP− 1 with a decrease in CLCR from 120 to 10 mL/min; however, a large 216

interindividual variability was noted. In addition, the diarrhea significantly decreased the AUC0-8 of both 217

MPA and AcMPAG in every CLCR, but did not affect the AUC0-8 of MPAG. The AUEC0-8 of IMPDH 218

with diarrhea was significantly higher than that without diarrhea in the case of CLCR under 60 mL/min. 219

The AUC0-8 of MPA significantly decreased with a reduction in serum albumin, although the 220

AUEC0–8 of IMPDH did not significantly change (Fig. 6). At a MPA concentration of 3.59 µg/mL, which 221

is equal to the population mean of IC50 in the case of CRP of 1.2 mg/dL, the IMPDH activity is 1.34-fold 222

15

higher in patients with CRP of 10 mg/dL, compared with that for CRP of 1.2 mg/dL. The AUEC0-8 of 223

IMPDH also significantly increases as the CRP rises. 224

225

Discussion 226

Patients undergoing HCT generally have intestinal mucosal damage due to a myeloablative or 227

reduced intensity conditioning regimen prior to HCT 33. Indeed, MPA concentrations in HCT patients are 228

generally lower than those of organ transplant patients despite an equivalent dose of MMF 34. In addition, 229

leukopenia and co-administered antibiotics induce the destruction of intestinal flora, leading to diarrhea. 230

The diarrhea decreased the reabsorption of MPA in the gastro-intestinal tract, and consequently decreased 231

the MPA concentration. In this study, IMPDH activity in CLCR under 60 mL/min with diarrhea was 232

significantly higher compared to those in the same CLCR without diarrhea in the same MPA dosing, 233

secondary to the PK changes. 234

In the early phase after transplantation, HCT patients suffer from renal impairment due to 235

thrombotic microangiopathy, which is an adverse effect caused by calcineurin inhibitors and high-dose 236

chemotherapy 35. Since MPA metabolites are excreted into the urine, the clearance of MPA metabolites 237

have been reported to decrease in association with lower renal function 14, 36. In the simulation using the 238

16

final PK/PD parameters, MPA concentration will be increased with a decreased CLCR, due to the 239

enhanced EHC of MPAG. Moreover, the IMPDH activity in CLCR under 30 mL/min was significantly 240

lower than that in 120 mL/min. Therefore, particular attention regarding extra-immunosuppression in 241

response to MPA is needed for patients with severe renal dysfunction. 242

The target range of MPA exposure may be influenced by changes in serum albumin, since the free 243

fraction of MPA was 1–2% 37. In the PD analysis, we speculated that the serum albumin had a significant 244

effect on the IC50 value of MPA due to the changed unbound fraction of MPA; however, we were unable 245

to conclusively demonstrate this effect due to a large inter- and intra-individual variability in IMPDH 246

activity. Therefore, changes in the serum albumin in clinical situations might not have a significant effect 247

on IMPDH activity, although it can affect the PK of MPA. 248

Interestingly, CRP exhibited a positive association with the IC50 for MPA. Patients undergoing HCT 249

suffered from various symptoms caused by an infection and/or the excessive production of inflammatory 250

cytokines on the days around the engraftment 38, 39. Indeed, the median of CRP in one week after starting 251

the MMF therapy (3.0 mg/dL) was significantly higher than that in three weeks after the therapy (0.8 252

mg/dL). Moreover, IMPDH1 is constitutively expressed in normal leukocytes, whereas IMPDH2 is 253

up-regulated in neoplastic and replicating cells40, 41. These findings suggest the reasons why CRP 254

17

exhibited a positive association with the IC50 for MPA. Whether the elevated CRP value reflects infection 255

or excessive production of inflammatory cytokines remains to be examined. 256

The simultaneous inclusion of IIVs and IOVs for PK/PD parameters significantly improved the 257

model fitting. HCT patients have single or multiple damages due to the conditioning regimen, infection or 258

excessive production of inflammatory cytokines33, 38, 39, 42. In addition, the recovery rate of organic and 259

hematopoietic functions following HCT showed a large variability. Therefore, the IIV and IOV in the 260

PK/PD of MPA in HCT patients should be large. 261

The present study included some limitations. The PK of MPA is usually expressed as 262

2-compartment model, and/or sometimes includes more sophisticated models 26, 27. In this study, the 263

model fitting was not improved by using any sophisticated models. The examined model largely depends 264

on the experimental design, and we would like to pick up clinical significant covariates on the PK of 265

MPA in a routine clinical care as shown in the previous our study 8. Additionally, although our population 266

size was modest, the estimated parameters, such as the ratio of EHC of MPAG or the IC50 for MPA, were 267

similar to those of previous reports 14. Therefore, the constructed PK/PD model for MPA was considered 268

to be appropriate. The effects of covariates extracted in the present study should be examined by 269

2-compartment model using more rich sampling data in a future. 270

18

In conclusion, we successfully constructed a population PK/PD model of MPA in patients 271

undergoing HCT. Renal dysfunction, diarrhea, and CRP are clinically significant factors affecting the PD 272

of MPA in the same dosing regimen. Dosage adjustment based on plasma MPA concentration is required 273

especially for patients with renal dysfunction and/or diarrhea. 274

275

Acknowledgments: The authors would like to thank Ms. Machiko Kaneyoshi for her assistant with 276

blood sampling. 277

278

References 279

1. Kaufman DB, Shapiro R, Lucey MR, Cherikh WS, Bustami RT, Dyke DB. Immunosuppression: 280

practice and trends. Am J Transplant 2004; 4: 38-53. 281

2. Minagawa K, Yamamori M, Katayama Y, Matsui T. Mycophenolate mofetil: fully utilizing its 282

benefits for GvHD prophylaxis. Int J Hematol 2012; 96: 10-25. 283

3. Dupuis R, Yuen A, Innocenti F. The influence of UGT polymorphisms as biomarkers in solid 284

organ transplantation. Clinica Chimica Acta 2012; 413: 1318-1325. 285

19

4. Wieland E, Shipkova M, Schellhaas U, Schutz E, Niedmann PD, Armstrong VW et al. Induction 286

of cytokine release by the acyl glucuronide of mycophenolic acid: a link to side effects? Clin 287

Biochem 2000; 33: 107-113. 288

5. Patel CG, Ogasawara K, Akhlaghi F. Mycophenolic acid glucuronide is transported by 289

multidrug resistance-associated protein 2 and this transport is not inhibited by cyclosporine, 290

tacrolimus or sirolimus. Xenobiotica 2013; 43: 229-235. 291

6. van Gelder T, Hilbrands LB, Vanrenterghem Y, Weimar W, de Fijter JW, Squifflet JP et al. A 292

randomized double-blind, multicenter plasma concentration controlled study of the safety and 293

efficacy of oral mycophenolate mofetil for the prevention of acute rejection after kidney 294

transplantation. Transplantation 1999; 68: 261-266. 295

7. Staatz CE, Tett SE. Clinical pharmacokinetics and pharmacodynamics of mycophenolate in solid 296

organ transplant recipients. Clin Pharmacokinet 2007; 46: 13-58. 297

8. Arai Y, Kondo T, Kitano T, Hishizawa M, Yamashita K, Kadowaki N et al. Monitoring 298

mycophenolate mofetil is necessary for the effective prophylaxis of acute GVHD after cord 299

blood transplantation. Bone Marrow Transplant 2015; 50: 312-314. 300

20

9. Wakahashi K, Yamamori M, Minagawa K, Ishii S, Nishikawa S, Shimoyama M et al. 301

Pharmacokinetics-based optimal dose prediction of donor source-dependent response to 302

mycophenolate mofetil in unrelated hematopoietic cell transplantation. Int J Hematol 2011; 94: 303

193-202. 304

10. Allison AC, Eugui EM. Mycophenolate mofetil and its mechanisms of action. 305

Immunopharmacology 2000; 47: 85-118. 306

11. Collart FR, Huberman E. Cloning and sequence-analysis of the human and chinese-hamster 307

inosine-5'-monophosphate dehydrogenase cdnas. J Biol Chem 1988; 263: 15769-15772. 308

12. Natsumeda Y, Ohno S, Kawasaki H, Konno Y, Weber G, Suzuki K. Two distinct cDNAs for 309

human IMP dehydrogenase. J Biol Chem 1990; 265: 5292-5295. 310

13. Carr SF, Papp E, Wu JC, Natsumeda Y. Characterization of human type-I and Type-II IMP 311

dehydrogenases. J Biol Chem 1993; 268: 27286-27290. 312

14. Li H, Mager DE, Sandmaier BM, Storer BE, Boeckh MJ, Bemer MJ et al. Pharmacokinetic and 313

pharmacodynamic analysis of inosine monophosphate dehydrogenase activity in hematopoietic 314

cell transplantation recipients treated with mycophenolate mofetil. Biol Blood Marrow 315

Transplant 2014; 20: 1121-1129. 316

21

15. de Winter BCM, Mathot RAA, Sombogaard F, Vulto AG, van Gelder T. Nonlinear relationship 317

between mycophenolate mofetil dose and mycophenolic acid exposure: implications for 318

therapeutic drug monitoring. Clin J Am Soc Nephrol 2011; 6: 656-663. 319

16. Frymoyer A, Verotta D, Jacobson PA, Long-Boyle J. Population pharmacokinetics of unbound 320

mycophenolic acid in adult allogeneic hematopoietic cell transplantation: effect of 321

pharmacogenetic factors. Br J Clin Pharmacol 2013; 75: 463-475. 322

17. Li H, Mager DE, Sandmaier BM, Maloney DG, Bemer MJ, McCune JS. Population 323

pharmacokinetics and dose optimization of mycophenolic acid in HCT recipients receiving oral 324

mycophenolate mofetil. J Clin Pharmacol 2013; 53: 393-402. 325

18. Barau C, Furlan V, Debray D, Taburet AM, Barrail-Tran A. Population pharmacokinetics of 326

mycophenolic acid and dose optimization with limited sampling strategy in liver transplant 327

children. Br J Clin Pharmacol 2012; 74: 515-524. 328

19. Kim H, Long-Boyle J, Rydholm N, Orchard PJ, Tolar J, Smith AR et al. Population 329

pharmacokinetics of unbound mycophenolic acid in pediatric and young adult patients 330

undergoing allogeneic hematopoietic cell transplantation. J Clin Pharmacol 2012; 52: 331

1665-1675. 332

22

20. Wang J, Yang JW, Zeevi A, Webber SA, Girnita DM, Selby R et al. IMPDH1 gene 333

polymorphisms and association with acute rejection in renal transplant patients. Clin Pharmacol 334

Ther 2008; 83: 711-717. 335

21. Sombogaard F, van Schaik RHN, Mathot RA, Budde K, van der Werf M, Vulto AG et al. 336

Interpatient variability in IMPDH activity in MMF-treated renal transplant patients is correlated 337

with IMPDH type II 3757T > C polymorphism. Pharmacogenet Genom 2009; 19: 626-634. 338

22. Naesens M, Kuypers DR, Verbeke K, Vanrenterghem Y. Multidrug resistance protein 2 genetic 339

polymorphisms influence mycophenolic acid exposure in renal allograft recipients. 340

Transplantation 2006; 82: 1074-1084. 341

23. Djebli N, Picard N, Rerolle JP, Le Meur Y, Marquet P. Influence of the UGT2B7 promoter 342

region and exon 2 polymorphisms and comedications on acyl-MPAG production in vitro and in 343

adult renal transplant patients. Pharmacogenet Genom 2007; 17: 321-330. 344

24. Kawanishi M, Yano I, Yoshimura K, Yamamoto T, Hashi S, Masuda S et al. Sensitive and 345

validated LC-MS/MS methods to evaluate mycophenolic acid pharmacokinetics and 346

pharmacodynamics in hematopoietic stem cell transplant patients. Biomed Chromatogr 2015; 347

29: 1309-1316. 348

23

25. Savic RM, Jonker DM, Kerbusch T, Karlsson MO. Implementation of a transit compartment 349

model for describing drug absorption in pharmacokinetic studies. J Pharmacokinet Pharacodyn 350

2007; 34: 711-726. 351

26. Jiao Z, Ding JJ, Shen J, Liang HQ, Zhong LJ, Wang Y et al. Population pharmacokinetic 352

modelling for enterohepatic circulation of mycophenolic acid in healthy Chinese and the 353

influence of polymorphisms in UGT1A9. Br J Clin Pharmacol 2008; 65: 893-907. 354

27. Funaki T. Enterohepatic circulation model for population pharmacokinetic analysis. J Pharm 355

Pharmacol 1999; 51: 1143-1148. 356

28. Karlsson MO, Sheiner LB. The importance of modeling interoccasion variability in population 357

pharmacokinetic analyses. J Pharmacokinet Biopharm 1993; 21: 735-750. 358

29. Yoshimura K, Yano I, Kawanishi M, Nakagawa S, Yonezawa A, Matsubara K. 359

Pharmacokinetics and pharmacodynamics of mycophenolic acid in Nagase analbuminemic rats: 360

Evaluation of protein binding effects using the modeling and simulation approach. Drug Metab 361

Pharmacoket 2015; 30: 441-448. 362

24

30. Upton RN, Mould DR. Basic concepts in population modeling, simulation, and model-based 363

drug development: part 3-introduction to pharmacodynamic modeling methods. CPT 364

Pharmacometrics Syst Pharmacol 2014; 3: e88. 365

31. Bergstrand M, Hooker AC, Wallin JE, Karlsson MO. Prediction-corrected visual predictive 366

checks for diagnosing nonlinear mixed-effects models. AAPS J 2011; 13: 143-151. 367

32. Beal SL. Ways to fit a PK model with some data below the quantification limit. J Pharmacokinet 368

Pharmacodyn 2001; 28: 481-504. 369

33. Chaudhry HM, Bruce AJ, Wolf RC, Litzow MR, Hogan WJ, Patnaik MS et al. The Incidence and 370

Severity of Oral Mucositis among Allogeneic Hematopoietic Stem Cell Transplantation Patients: 371

A Systematic Review. Biol Blood Marrow Transplant 2016; 22: 605-616. 372

34. van Hest RM, Doorduijn JK, de Winter BC, Cornelissen JJ, Vulto AG, Oellerich M et al. 373

Pharmacokinetics of mycophenolate mofetil in hematopoietic stem cell transplant recipients. 374

Ther Drug Monit 2007; 29: 353-360. 375

35. Kemmner S, Verbeek M, Heemann U. Renal dysfunction following bone marrow transplantation. 376

J Nephrol. 2017; 30: 201-209. 377

25

36. Sam WJ, Akhlaghi F, Rosenbaum SE. Population pharmacokinetics of mycophenolic acid and 378

its 2 glucuronidated metabolites in kidney transplant recipients. J Clin Pharmacol 2009; 49: 379

185-195. 380

37. Nowak I, Shaw LM. Mycophenolic-acid binding to human serum-albumin - characterization and 381

relation to pharmacodynamics. Clin Chem 1995; 41: 1011-1017. 382

38. Cornell RF, Hari P, Drobyski WR. Engraftment syndrome after autologous stem cell 383

transplantation: an update unifying the definition and management approach. Biol Blood Marrow 384

Transplant 2015; 21: 2061-2068 385

39. Sahin U, Toprak SK, Atilla PA, Atilla E, Demirer T. An overview of infectious complications 386

after allogeneic hematopoietic stem cell transplantation. J Infect Chemother 2016; 22: 505-514 387

40. Nagai M, Natsumeda Y, Weber G. Proliferation-linked regulation of type-II IMP dehydrogenase 388

gene in human normal lymphocytes and HL-60 leukemic-cells. Cancer Res 1992; 52: 258-261. 389

41. Nagai M, Natsumeda Y, Konno Y, Hoffman R, Irino S, Weber G. Selective up-regulation of 390

type-II Inosine 5'-monophosphate dehydrogenase messenger RNA expression in human 391

leukemias. Cancer Res 1991; 51: 3886-3890. 392

26

42. Uchida N, Wake A, Nakano N, Ishiwata K, Takagi S, Tsuji M et al. Mycophenolate and 393

tacrolimus for graft-versus-host disease prophylaxis for elderly after cord blood transplantation: 394

a matched pair comparison with tacrolimus alone. Transplantation. 2011; 92: 366-371. 395

396

27

Figure legends 397

Fig. 1 Overview of the basic pharmacokinetic and pharmacodynamic model characterizing mycophenolic 398

acid (MPA), MPA glucuronide (MPAG), MPA acylglucuronide (AcMPAG), and 399

inosine-5’-monophosphate dehydrogenase (IMPDH). GI, gastrointestinal tract; Ka, first-order absorption 400

rate constant; CLMPA, clearance of MPA; CLMPAG, clearance of MPAG; CLAcMPAG, clearance of 401

AcMPAG; VMPA, volume of distribution of MPA; VMPAG, volume of distribution of MPAG; VAcMPAG, 402

volume of distribution of AcMPAG; KEHC, first-order rate constant of enterohepatic circulation; FM1, 403

fraction of MPA converted to MPAG. 404

405

Fig. 2 Interoccasional pharmacokinetic and pharmacodynamic parameters between one and three weeks 406

after initiation of mycophenolate mofetil (MMF) therapy. (A) The first-order absorption rate constant 407

(Ka); (B) relative bioavailability (F); (C) clearance of MPAG (CLMPAG); (D) clearance of AcMPAG 408

(CLAcMPAG); (E) first-order rate constant for the enterohepatic circulation (KEHC); (F) baseline IMPDH 409

activity (E0). 410

411

28

Fig. 3 Goodness-of-fit plots of the observed versus population predictions (A-D) or individual predictions 412

(E-H) using the final model. (A and E) MPA concentrations; (B and F) MPAG concentrations; (C and G) 413

AcMPAG concentrations; (D and H) IMPDH activity. Each dotted line denotes the line of identity. 414

415

Fig. 4 Prediction corrected visual predictive check plots. All open circles represent the observed 416

concentrations or IMPDH activities (prediction corrected). (A-D) one week after initiation of MMF 417

therapy; (E-H) three weeks after initiation of MMF therapy. The solid line represents the median of the 418

observed data. The dotted line represents the observed 5th and 95th percentiles. The shaded area denotes 419

the simulation-based 95% confidence interval for the median or the 5th and 95th percentiles. 420

421

Fig. 5 Simulation for the effects of creatinine clearance (CLCR) and diarrhea on the pharmacokinetics and 422

pharmacodynamics of MPA in typical patients based on the final population model. (A) AUC0-8 of MPA; 423

(B) AUC0-8 of MPAG; (C) AUC0-8 of AcMPAG; (D) AUEC0–8 of IMPDH activity. The dose of MMF 424

was fixed to 500 mg every 8 h. Each box plot represents the 5th percentile, lower quartile, median, upper 425

quartile, and 95th percentile values obtained from 1000 simulated data sets. *; p < 0.05; ***; p < 0.001, 426

significantly different from the group with a CLCR of 120 mL/min by the Kruskal-waillis test following 427

29

by the Dunn test. †; p < 0.05;†††; p < 0.001, significantly different from the same CLCR without diarrhea 428

by the Kruskal-waillis test following by the Dunn test. 429

430

Fig. 6 Simulation for the effects of serum albumin (A and B) and C-reactive protein (CRP) (C and D) on 431

the pharmacokinetics and pharmacodynamics of MPA in typical patients based on the final population 432

model. The dose of MMF was fixed to 500 mg every 8 h. (A) AUC0-8 of MPA; (B) AUEC0-8 of IMPDH 433

activity; (C) the relationship between the MPA plasma concentration and IMPDH activity. The dotted, 434

thin, and thick lines represent 0.1, 1.2, and 10 mg/dL of CRP, respectively. The vertical line represents 435

the MPA concentration of 3.59 µg/mL (the population mean of IC50 in the case of CRP 1.2 mg/dL); (D) 436

AUEC0-8 of IMPDH activity. Each box plot represents the 5th percentile, lower quartile, median, upper 437

quartile, and 95th percentile values obtained from 1000 simulated data sets. ***; p < 0.001, significantly 438

different from the group of 4.2 mg/dL of serum albumin by the Kruskal-waillis test following by the 439

Dunn test. †††; p < 0.001, significantly different from the group of 0.1 mg/dL of CRP by the 440

Kruskal-waillis test following by the Dunn test. 441

Fig. 1

MPACentralVMPA

MPAGCentralVMPAG

AcMPAGCentralVAcMPAG

GI

IMPDH

Ka

KEHC

FM1×CLMPA

(1-FM1)×CLMPACLMPAG

CLAcMPAG

DOSE

!

! !

Fig. 2

1 week 3 week0

2

4

6

Ka (h

-1)

1 week 3 week0.0

0.5

1.0

1.5

2.0

2.5

F

1 week 3 week0

1

2

3

4

5

CL M

PAG

(L/h

)

1 week 3 week0

200

400

600

800

1000

E 0(µ

mol·s

ec-1·m

ol A

MP-1

)

1 week 3 week0

2

4

6

8

CL Ac

MPA

G (L

/h)

(A) (B) (C)

(D) (E)

1 week 3 week0.0

0.1

0.2

0.3

0.4K EH

C (h

-1)

(F)

O>-------;Q

Fig. 3

0 5 10 150

5

10

15

MPA (µg/mL)

MPA

(µg/

mL)

0 5 10 150

5

10

15

MPA (µg/mL)

MPA

(µg/

mL)

0 50 100 150 2000

50

100

150

200

MPAG (µg/mL)M

PAG

(µg/

mL)

0 50 100 150 2000

50

100

150

200

MPAG (µg/mL)

MPA

G (µ

g/m

L)0 1 2 3

0

1

2

3

AcMPAG (µg/mL)

AcM

PAG

(µg/

mL)

0.1 1 10 10010000.1

1

10

100

1000

IMPDH Activity(µmol·sec -1·mol AMP -1)

IMPD

H A

ctiv

ity(µ

mol·s

ec -1·m

ol A

MP

-1)

0.1 1 10 10010000.1

1

10

100

1000

IMPDH Activity(µmol·sec -1·mol AMP -1)

IMPD

H A

ctiv

ity(µ

mol·s

ec -1·m

ol A

MP

-1)

Population prediction

Individual prediction

0 1 2 30

1

2

3

AcMPAG (µg/mL)

AcM

PAG

(µg/

mL)

Obs

erve

d(A) (B) (C) (D)

(E) (F) (G) (H)

0

0 0

8 0

80 0

0

0 0

0

,t, 00

0

o0

0

0 0

0

0

o0

0 O 00

8 0

0 00 o 0

0 000

0 0

o oo oo//./ 0 0 .. •····

00 O.·/

OO O O O •.•.• ·•··

'\,o0 ... •· ... •· 0

'i, ./ '// .. •·············t .......... 80

.. / Oo"

... •··············· 0

J./

~: 0.vi

.9-·!1···, 0 8 o./ 0 o

o .. l) o ..... Q90

.. •················ 0

Fig. 4

0 4 8 120

5

10

15

20

MPA

(µg/

mL)

0 4 8 120

50

100

150

MPA

G (µ

g/m

L)

0 4 8 120

1

2

3

4

5

AcM

PAG

(µg/

mL)

0 4 8 120

5

10

15

20

MPA

(µg/

mL)

0 4 8 120

50

100

150

MPA

G (µ

g/m

L)

0 4 8 120

1

2

3

4

5

AcM

PAG

(µg/

mL)

0 4 8 121

10

100

1000

IMPD

H a

ctiv

ity(µ

mol·s

ec -1·m

ol A

MP

-1)

(A) (B) (C) (D)

(E) (F) (G) (H)

Time after dosing (h)

0 4 8 121

10

100

1000

IMPD

H a

ctiv

ity(µ

mol·s

ec -1·m

ol A

MP

-1)

0 0 0

0

0 @

0

0

0

"'- 0

0 0

8 --,,_ __ .., _______ _

!l ! J..r-;~ ;-~ -J § 0 §

------s ·----%-----------------0----0 0 0

8

0 0 0

0 0

0

0

G/,--'\,,, 0

0

• 0 0

Fig. 5

120 90 60 30 10 12

0 90 60 30 100

50

100

150

CLCR (mL/min)

MPA

AU

C0-

8 (µ

g·h·

mL-1

)

Diarrhea( - ) (+)

***

******

***

†††

***

*** ***

†††

†††††††††

120 90 60 30 10 12

0 90 60 30 100

40

80

120

CLCR (mL/min)

AcM

PAG

AU

C0-

8 (µ

g·h·

mL-1

)

*********

***

Diarrhea( - ) (+)

*********

***†††

††††††††††††

120 90 60 30 10 12

0 90 60 30 100

1000

2000

3000

CLCR (mL/min)

MPA

G A

UC

0-8

(µg·

h·m

L-1)

***

******

***

Diarrhea( - ) (+)

***

******

***

120 90 60 30 10 12

0 90 60 30 100

500

1000

1500

2000

CLCR (mL/min)

IMPD

H A

UEC

0-8

(µm

ol·h·s

ec -1·m

ol A

MP

-1)

******

††††††

Diarrhea( - ) (+)

***

(A) (B)

(C) (D)

ffl-t

t{I]

----

---i

t-IT

:J--

----

--­

t-IT

:J--

----

--­

i-(I

J---

----

---

~

~

t-C

CJ-

----

----

­t-

CC

J---

----

--­

t-C

CJ-

----

----

­t-

IT:J

----

----

­i-

[IJ-

----

----

-

l{]]--

-i

fil--

1 t{

[}--

--i

t{I]

----

----

t

~

l{Il--

l t{I

l--1

l{Il-

t l[}

---i

ID--

----

-,

t[}-i

1{[1--

1 @

--,

t-{I

]---

--i

~

t[}-i

1{[1--

1 @

--,

t-{I

]---

--i

~

Fig. 6

4.2 3.2 2.20

10

20

30

40

Albumin (g/dL)

MPA

AU

C0-

8 (µ

g·h·

mL-1

)

***

***

0 4 8 120

20

40

60

80

MPA concentration (µg/mL)

IMPD

H A

ctiv

ity(µ

mol·s

ec -1·m

ol A

MP

-1) 0.1

1.210

4.2 3.2 2.20

500

1000

1500

2000

Albumin (g/dL)

IMPD

H A

UEC

0-8

(µm

ol·h·s

ec -1·m

ol A

MP

-1)

0.1 1.2 100

500

1000

1500

2000

CRP (mg/dL)

IMPD

H A

UEC

0-8

(µm

ol·h·s

ec -1·m

ol A

MP

-1) †††

†††

(A) (B)

(C) (D)

··• ................................ .

Table 1. Characteristics of patients included in the analysis

Characteristics Number or median (range)

Gender (male/female) 34/15

Stem cell source (CBT/BMT/PBT) 34/13/2

RIC/MAC 23/26

Body weight (kg) 61.9 (33.5–84.2)

Age (years) 49 (21–66)

AST (U/L) 18 (6–206)

Serum albumin (mg/dL) 3.2 (1.6–4.1)

Total bilirubin (mg/dL) 0.5 (0.2–2.1)

Creatinine clearance (mL/min) 112 (20–262)

C-reactive protein (mg/dL) 1.2 (0–25.1)

Dose of MMF (mg/day) 1750 (1000–3000)

Dose normalized by body weight

(mg/day/kg)

30.3 (21.3–57.7)

Diarrhea 1 and 3 weeks after MMF therapy 29/25

Genetic polymorphisms

(wild/heterozygote/homozygote)

Pre-transplant a Post-transplant

MRP2 −24 C>T (rs717620) 35/12/0 -

UGT2B7 −842G>A (rs7439366) 20/21/6 -

IMPDH1 −106G>A (rs2278294) 12/23/12 11/23/15

IMPDH1 125G>A (rs2278293) 18/20/9 20/18/11

IMPDH2 3757T>C (rs11706052) 44/2/1 41/8/0

aGenotypes in two patients were unknown because one patient underwent the second-stem cell

transplantation and the other patient was defective in collecting recipient’s DNA.

CBT, cord blood transplantation; BMT, bone marrow stem cell transplantation; PBT, peripheral blood

stem cell transplantation; RIC, reduced-intesity conditioning, MAC, myeloablative conditioning; AST,

aspartate aminotransferase; MMF, mycophenolate mofetil; MRP, multidrug resistance-associated protein;

UGT, UDP-glucuronosyltransferase; IMPDH, inosine-5’-monophosphate dehydrogenase

Table 2. Final population pharmacokinetic and pharmacodynamic parameters of MPA

Parameters Mean RSE (%)

Pharmacokinetic parameters

Ka (h−1) 1.18 39.5

F 1 FIX -

θCL,MPA (L/h) 44.5 17.0

θV,MPA (L) 41.5 17.3

θCL,MPAG (L/h) 2.46 14.8

VMPAG (L) 17.3 17.1

θK,EHC (h-1) 0.0635 23.8

θCL,AcMPAG (L/h) 3.17 16.7

VAcMPAG (L) 5.50 15.7

Covariates

Diarrhea on KEHC 0.375 30.7

CLCR on CLMPAG 0.730 69.7

CLCR on CLAcMPAG 0.537 17.0

ALB on VMPA −3.71 29.4

ALB on CLMPA −1.02 43.2

Interindividual variability (IIV; %)

VMPA 56.9 38.3

F 84.9 6.06

Interoccasional variability (IOV; %)

Ka 97.9 24.6

F 19.1 47.7

CLMPAG 33.0 22.7

KEHC 91.2 21.9

CLAcMPAG 42.0 23.8

Residual variability (proportional error; %)

MPA concentration 40.9 12.4

MPAG concentration 18.8 10.5

AcMPAG concentration 31.4 6.53

Pharmacodynamic parameters

E0 (µmol·h·sec−1·mol AMP−1) 65.5 10.4

θIC50 (µg/mL) 3.59 20.3

Covariates

CRP on IC50 0.338 47.3

Interindividual variability (IIV, %)

IC50 81.2 45.3

Interoccasional variability (IOV, %)

E0 85.8 27.4

Residual variability (proportional error; %)

IMPDH activity 46.6 3.73

Ka, first-order absorption rate constant; F, relative bioavailability; CLMPA, clearance of MPA; VMPA,

volume of distribution of MPA; CLMPAG, clearance of MPAG; VMPAG, volume of distribution of MPAG;

KEHC, first-order rate constant of enterohepatic circulation; CLAcMPAG, clearance of AcMPAG; VAcMPAG,

volume of distribution of AcMPAG; CLCR, creatinine clearance; ALB, serum albumin concentration; E0,

baseline of IMPDH activity; IC50, half-maximal inhibitory concentration; CRP, C-reactive protein.

The final pharmacokinetic and pharmacodynamic models were described as followed: CL

θ , .

. ; V θ , . . , CL θ , . ,

K θ , 0.375 , CL θ , . , and IC θ

.. ; COV is set to 0 for patients without diarrhea and 1 for patients with diarrhea.