You are on page 1of 11

Nephrol Dial Transplant (2020) 35: 1060–1070

doi: 10.1093/ndt/gfaa111

Effect of the proton-pump Inhibitor pantoprazole on


MycoPhenolic ACid exposure in kidney and liver transplant
ORIGINAL ARTICLE

recipienTs (IMPACT study): a randomized trial

Andrew Sunderland1, Graeme Russ2, Benedetta Sallustio3,4, Matthew Cervelli5, David Joyce6,7,8,

Downloaded from https://academic.oup.com/ndt/article/35/6/1060/5855235 by guest on 06 November 2023


Esther Ooi6,8, Gary Jeffrey8,9, Neil Boudville1,8, Aron Chakera1, Gursharan Dogra1, Doris Chan1,
Germaine Wong10,11,12 and Wai H. Lim1,8
1
Department of Renal Medicine, Sir Charles Gairdner Hospital, Perth, Australia, 2Department of Nephrology & Transplantation Services,
Royal Adelaide Hospital, and University of Adelaide, Adelaide, Australia, 3Discipline of Pharmacology, Adelaide Medical School, The University
of Adelaide, Adelaide, Australia, 4Department of Clinical Pharmacology, The Queen Elizabeth Hospital, Adelaide, Australia, 5Department of
Pharmacy, Royal Adelaide Hospital, Adelaide, Australia, 6School of Biomedical Sciences, University of Western Australia, Perth, Australia,
7
Biochemistry and Toxicology, PathWest, Perth, Australia, 8Faculty of Health and Medical Sciences, Medical School, University of Western
Australia, Perth, Australia, 9Department of Hepatology Unit, Sir Charles Gairdner Hospital, Perth, Australia, 10Sydney School of Public Health,
University of Sydney, Sydney, Australia, 11Centre for Transplant and Renal Research, Westmead Hospital, Sydney, Australia and 12Centre for
Kidney Research, The Children’s Hospital at Westmead, Sydney, Australia

Correspondence to: Wai H. Lim; E-mail: wai.lim@health.wa.gov.au

GRAPHICAL ABSTRACT

In kidney and liver transplant patients, co-administration of pantoprazole reduces


RCT bioavailability of mycophenolic acid from mycophenolate mofetil (MMF), but
increases bioavailability from enteric-coated mycophenolic acid salts (EC-MPS)

Trial characteristics Trial design Primary outcome


2 centres (Australia) Placebo Placebo Placebo
n = 40 participants n=9 n = 10 53.9
MMF
Age > 18 years PPI PPI MPA–AUC PPI
Kidney/liver transplant > 6 months
(n = 19) mg*hr/L 43.8
n = 10 n=9
P = 0.004
Current medication:
Treatment Washout Treatment Bioavailabilty of
MMF ≥ 1 g daily or
7 days 7 days 7 days
EC-MPS ≥ 1080 mg daily mycophenolic acid
Intervention: Placebo Placebo Placebo
Pantoprazole 40 mg daily n = 11 n = 10 36.1
Primary endpoint: EC-MPS MPA–AUC PPI
PPI PPI
12 hr bioavailability of (n = 21) n = 10 n = 11 mg*hr/L 45.9
mycophenolic acid (MPA–AUC) P = 0.023

Sunderland A, et al. NDT (2020)


@NDTSocial

ABSTRACT concentrations and adverse allograft outcome. Proton-pump


Background. Mycophenolic acid (MPA) is widely utilized as an inhibitors (PPIs) may have variable effects on the absorption of
immunosuppressant in kidney and liver transplantation, with different MPA formulations leading to differences in MPA
reports suggesting an independent relationship between MPA exposure.
C The Author(s) 2020. Published by Oxford University Press on behalf of ERA-EDTA. All rights reserved.
V 1060
Methods. A multicentre, randomized, prospective, double- KEY LEARNING POINTS
blind placebo-controlled cross-over study was conducted to de-
termine the effect of the PPI pantoprazole on the MPA and its
metabolite MPA-glucuronide (MPA-G) area under the curve What is already known about this subject?
(AUC) >12 h (MPA-AUC12 h) in recipients maintained on • Proton-pump inhibitors (PPIs) could reduce the sys-
mycophenolate mofetil (MMF) or enteric-coated mycopheno- temic exposure of mycophenolic acid (MPA) for
late sodium (EC-MPS). We planned a priori to examine sepa- patients maintained on mycophenolate mofetil (MMF)
rately recipients maintained on MMF and EC-MPS for each formulation but not in those maintained on enteric-
pharmacokinetic parameter. The trial (and protocol) was regis- coated salt form mycophenolate sodium (EC-MPS),
tered with the Australian New Zealand Clinical Trials Registry but this drug–drug interaction has not been confirmed
on 24 March 2011, with the registration number of in adequately powered randomized controlled trial in
ACTRN12611000316909 (‘IMPACT’ study). solid organ transplant recipients.
Results. Of the 45 recipients screened, 40 (19 MMF and 21 EC- What this study adds?
MPS) were randomized. The mean (standard deviation) recipient • This randomized placebo-controlled cross-over study

Downloaded from https://academic.oup.com/ndt/article/35/6/1060/5855235 by guest on 06 November 2023


age was 58 (11) years with a median (interquartile range) time showed that the co-administration of pantoprazole
post-transplant of 43 (20–132) months. For recipients on MMF, substantially reduced the bioavailability of MPA in
there was a significant reduction in the MPA-AUC12 h [geometric kidney and liver transplant recipients maintained on
mean (95% confidence interval) placebo: 53.9 (44.0–65.9) mg*h/ MMF and had the opposite effect in patients main-
L versus pantoprazole: 43.8 (35.6–53.4) mg*h/L; P ¼ 0.004] when tained on EC-MPS.
pantoprazole was co-administered compared with placebo. In What impact this may have on practice or policy?
contrast, co-administration with pantoprazole significantly in- • Clinicians should be cognizant of this drug interaction
creased MPA-AUC12 h [placebo: 36.1 (26.5–49.2) mg*h/L versus when prescribing PPIs with MMF or EC-MPS, but the
pantoprazole: 45.9 (35.5–59.3) mg*h/L; P ¼ 0.023] in those re- longer term clinical effect of this drug–drug interac-
ceiving EC-MPS. Pantoprazole had no effect on the pharmacoki- tion in kidney and liver transplant recipients remains
netic profiles of MPA-G for either group. unknown.
Conclusions. The co-administration of pantoprazole substantially
reduced the bioavailability of MPA in patients maintained on
MMF and had the opposite effect in patients maintained on EC-
MPS, and therefore, clinicians should be cognizant of this drug in- oesophageal reflux disease [6]. It has been shown that by increas-
teraction when prescribing the different MPA formulations. ing gastric pH and thus reducing the solubility of MMF in the
Keywords: drug interaction, enteric-coated mycophenolate gut, PPIs could reduce the systemic exposure of MPA for patients
sodium, kidney transplant, mycophenolate, mycophenolic on MMF but not EC-MPS [7–9]. This effect may lead to inade-
acid, pharmacokinetic, proton-pump inhibitor quate immunosuppression with an increased risk of rejection.
This important effect of PPIs on MPA pharmacokinetics follow-
ing MMF or EC-MPS administration remains poorly recognized
and should be taken into consideration with regard to appropri-
INTRODUCTION ate dosing of MMF and EC-MPS in kidney and liver transplant
Mycophenolic acid (MPA) is available as the ester pro-drug recipients. In a single-dose cross-over study of 12 healthy volun-
R
mycophenolate mofetil (MMF, CellceptV) or as the enteric- teers, the concomitant administration of pantoprazole signifi-
coated salt form mycophenolate sodium (EC-MPS, MyforticV).
R
cantly reduced MPA exposure following MMF treatment (dose
Immunosuppressive regimens containing MPA are widely uti- 1 g daily), which was not evident following EC-MPS (720 mg
lized in kidney and liver transplantation to reduce rejection daily) treatment [10]. In stable kidney and liver transplant recipi-
rates and improve allograft survival [1]. There has been consid- ents, we aimed to compare the effects of a PPI versus placebo on
erable interest in individualizing MMF or EC-MPS dosing MPA exposure in those maintained on MMF or EC-MPS.
according to therapeutic drug monitoring (TDM) to reduce ad-
verse events associated with these drugs while improving clini-
cal outcomes. The APOMYGRE and OPTICEPT trials suggest MATERIALS AND METHODS
that adjustment of MMF dosing according to therapeutic MPA This was a multicentre, randomized, prospective, double-
monitoring could reduce the risk of treatment failure, including blinded, placebo-controlled, cross-over study to assess the effect
rejection, but the impact of MPA monitoring on longer term al- of gastric acid suppression (PPI pantoprazole) on the pharma-
lograft outcome remains unclear [2–5]. Consequently, MPA cokinetics of MPA in kidney or liver transplant recipients main-
TDM has not been widely adopted into clinical practice among tained on MMF or EC-MPS. This trial was conducted in two
transplanting centres. transplanting centres in Australia (Sir Charles Gairdner
Peptic ulcer disease is a common complication following kid- Hospital, Perth; Royal Adelaide Hospital (RAH), Adelaide].
ney and liver transplantation and therefore prophylactic proton- Local institutional ethics committees at each site approved the
pump inhibitors (PPIs) are often prescribed in these patients to study and written consents were obtained from participants
reduce the risk of symptomatic peptic ulcer or gastro- prior to randomization. The trial (and protocol) was registered

Effect of pantoprazole on MPA exposure 1061


with the Australian New Zealand Clinical Trials Registry on 24 Professionals (Victoria, Australia) on the advice of the clinical
March 2011, with the registration number of pharmacologist from RAH. The pantoprazole (commercial
ACTRN12611000316909 (‘IMPACT’ study). product) pills were de-blistered and placed into an empty gela-
Study population tine capsule and then filled with lactose or other inert filler to
disguise their content. The placebo was made by encapsulating
The target population comprised stable adult (>18 years) lactose in an identical soft gelatine capsule. Both capsules were
kidney and liver transplant recipients who were 6 months indistinguishable visually or by smell from the matching pla-
post-transplant. Recipients were maintained on MMF (1 g cebo, and dissolved completely once ingested. The trial pharma-
daily) or EC-MPS (1080 mg daily) in two equally divided cists at the RAH prepared two packs labelled Packs A and B,
doses. If recipients were maintained on unequal twice daily dos- with each pack containing seven tablets of pantoprazole or
ing, then they agreed to alter to two equally divided dose for matching placebo, with packs shipped to Perth. The package of
2 weeks prior to trial enrolment. Other inclusion criteria for the either study medication into Pack A or B was random, under-
trial were recipients who were willing to change to pantoprazole taken by the trial pharmacist of RAH. The selection of the pack-
(from other PPI or H2-blocker) or willing to start pantoprazole age of either study medication for each recipient was
if not already on this drug, maintained on a stable dose of calci-

Downloaded from https://academic.oup.com/ndt/article/35/6/1060/5855235 by guest on 06 November 2023


undertaken by the trial pharmacy team of each site independent
neurin inhibitor (CNI; cyclosporine or tacrolimus) and were ca- of the study according to a pre-specified allocation sequence.
pable of providing written informed consent. Exclusion criteria The study investigators, study coordinators and recipients were
included recipients maintained on non-CNI agents except cor- blinded to the study medications. There were 56 study medica-
ticosteroids; recent change (<2 weeks) in the total dose of corti- tion packs prepared, with each pack containing Packs A and B
costeroids, CNI, MMF or EC-MPS; recent acute rejection (16 packs were unused/expired and discarded). These packs
(defined as biopsy-proven acute rejection episode requiring in- were block randomized for the two sites and for liver and kid-
tervention within 1 month prior to randomization); ney transplant recipients.
Modification of Diet in Renal Disease Study-derived estimated
glomerular filtration rate (eGFR) of <20 mL/min/1.73 m2; and Clinical endpoints
significant peptic ulcer disease or ulcerative esophagitis (based The primary endpoint of this study was the pharmacokinetic
on medical history) where withdrawal of acid suppression ther- profile of MPA and MPA-glucuronide (MPA-G) when co-
apy is not clinically appropriate, and concurrent use of magne- administered with placebo or pantoprazole, expressed as
sium- or aluminium-based antacid or cholestyramine. MPA-area under the curve (AUC) and MPA-G AUC >12 h
Study design (MPA-AUC12 h and MPAG-AUC12 h, respectively), maximum
For recipients who were maintained on PPI and/or H2- concentration (Cmax) and time to Cmax (Tmax). Dose-
blocker, this was ceased 14 days prior to randomization. normalized AUC12 h and Cmax for MPA and MPA-G were cal-
Recipients were given two packets of study medications clearly culated for a 2 g daily dose of MMF or 1440 mg daily dose of
labelled Pack A and B. Recipients were advised to commence EC-MPS. Secondary endpoints included any biopsy-proven
study medication from Pack A (containing either placebo or acute rejection, allograft failure and adverse events (as assessed
pantoprazole, one tablet daily for 7 days; Days 1–7) and then by site investigators) during and for 2 weeks following the com-
admitted on the morning of the first pharmacokinetic study pletion of the study.
(Day 8). The previous dose of MMF or EC-MPS was taken 12 h
Measurements of MPA and MPA-G
prior to the first blood sample (Time 0). Following the initial ve-
nous blood sampling, the recipients took the morning dose of Plasma was analysed for MPA and MPA-G using a validated
MMF or EC-MPS, along with the CNI and other usual medica- high-performance liquid chromatography-ultraviolet-based
tions as prescribed. Venous blood samplings occurred at time method detection assay [11]. The area under the plasma MPA
point 0, and then 0.5, 1, 1.5, 2, 3, 4, 5, 6, 8, 10 and 12 h post and MPA-G concentration–time curves (i.e. AUC) to the last
MMF or EC-MPS dose. The blood samples were centrifuged quantifiable concentration (AUCt) were calculated using the
and plasma separated and stored at 70 C until analysis. log-trapezoidal rule. All assays were performed in a laboratory
Following the completion of the first visit, the recipients were accredited by the National Association of Testing Authorities,
advised to continue on the usual medications for the next 7 days Australia (The Queen Elizabeth Hospital, Adelaide, Australia)
without any study medications (washout period; Days 8–14). that subscribes to an external quality assurance programme for
On Day 15, the recipients were advised to commence study MPA. The intra-assay precision and accuracy for MPA were es-
medication from Pack B (containing either placebo or panto- timated at calibrator concentrations at 0.5 and 20 mg/L and for
prazole, one tablet daily for 7 days; Days 15–21), and then re- MPA-G at calibrator concentrations of 5 and 200 mg/L. Inter-
admitted on Day 22 for the second pharmacokinetic study. A assay precision and accuracy of quality control samples were
telephone call reminder was scheduled at Day 14 and a compli- determined for MPA at 2 and 15 mg/L and for MPA-G at 20
ance check of study medication was undertaken at each phar- and 150 mg/L. Coefficients of variation were <5.0% and biases
macokinetic study visit. were <14.0% for each concentration of each analyte.
Study drug Sample size calculation
The study medications of pantoprazole (40 mg) and match- The sample size calculation was derived from the randomized
ing placebo were prepared by Pharmaceutical Packaging cross-over study in healthy volunteers by Rupprecht et al. [10],

1062 A. Sunderland et al.


Total participants consented (n=45)
44 kidney transplant recipients + 1 liver transplant recipient

4 screen failures
and not randomized

Total participants randomized (n=41)


40 kidney transplant recipients + 1 liver transplant recipient

1 withdrew consent
(medication change)

MMF (n=19) EC–MPS (n=21)


18 kidney transplants 21 kidney transplants

Downloaded from https://academic.oup.com/ndt/article/35/6/1060/5855235 by guest on 06 November 2023


1 liver transplant 0 liver transplants

Period 1 Placebo (n=9) PPI (n=10) Placebo (n=11) PPI (n=10) Period 1
7 days 12h AUC 7 days 12h AUC

Washout period
(7 days)

Period 2 Placebo (n=10) PPI (n=9) Placebo (n=10) PPI (n=11) Period 2
7 days 12h AUC 7 days 12h AUC

FIGURE 1: The CONSORT flow diagram and trial design of the ‘IMPACT’ trial. Of 40 recipients who were randomized and had completed
R
the trial, 19 were maintained on MMF and 21 were maintained on EC-MPS (MyforticV). Recipients were randomized to the study medication
(i.e. placebo or PPI 40 mg of pantoprazole daily) for 7 days, followed by a wash-out period of 7 days and then the alternative study medication
for 7 days. Two 12-h pharmacokinetic profiles were undertaken at the completion of each study medication.

where 12 healthy subjects were sufficient to detect a significant pantoprazole for each pharmacokinetic parameter within each
difference between MPA AUC12 h following self-administration study group of MMF and EC-MPS. The proportions of recipi-
of MMF with and without pantoprazole (MMF 40.0 6 7.8 versus ents in each study group with MPA-AUC12 h with thresholds of
MMF þ pantoprazole 29.3 6 7.4 mg*h/L; P < 0.001). Given the >30, >40, >50 and >60 mg*h/L were determined, given the
effect size (of 25% reduction of MPA-AUC12 h) and the poten- MPA-AUC12 h threshold of 30–60 mg*h/L has been shown to
tial greater variability observed in this study, a sample size of 15 be associated with lower risk of acute rejection [2, 5, 12–14].
recipients maintained on MMF would be sufficient to detect a Two sensitivity analyses were undertaken: (i) excluding one
significant difference in MPA-AUC12 h of 25% with and with- liver transplant recipient (for the MMF group) and (ii) stratified
out pantoprazole. A similar number will be recruited for recipi- by entry eGFR thresholds of 60 and >60 mL/min/1.73 m2. All
ents maintained on EC-MPS. We planned to recruit 42 recipients analyses were undertaken using Stata version 15.1 (StataCorp
(similar proportion of recipients maintained on MMF and EC- LP, College Station, TX, USA), with P < 0.05 considered statis-
MPS), assuming a drop-out rate of up to 30%. tically significant.

Statistical analysis
Data were expressed as number (percentage), mean [stan- RESULTS
dard deviation (SD)] and median [interquartile range (IQR)] Forty-five liver and kidney transplant recipients were consented
for categorical, normally distributed and non-normally distrib- and 41 underwent randomization (four screen failures prior to
uted continuous variables, respectively, stratified by the study randomization) between December 2011 and August 2017.
groups. We planned a priori to examine separately recipients One recipient withdrew consent after randomization, resulting
maintained on MMF and EC-MPS, and between the subgroups in 40 recipients completing the study (39 kidney and 1 liver
of CNI types (cyclosporin and tacrolimus) for each pharmaco- transplant recipients). We were unable to recruit additional
kinetic parameter. For MPA and MPA-G (6dose-normalized) liver transplant recipients over the 5-year period and therefore
AUC12 h, Cmax and Tmax, the geometric means and 95% confi- had continued to recruit kidney transplant recipients until the
dence intervals (CIs) were derived, with Wilcoxon signed-rank target study sample size was reached. The CONSORT flow dia-
test used to test the difference between placebo versus gram for this trial is shown in Figure 1.

Effect of pantoprazole on MPA exposure 1063


Table 1. Baseline characteristics of study participants

Baseline characteristics of study cohort MMF (n ¼ 19) EC-MPS (n ¼ 21) Total (n ¼ 40)
Age, mean (SD) 61.1 (28.9) 55.8 (12.3) 58.3 (11.0)
Gender
Male 10 (52.6) 17 (81.0) 27 (67.5)
Female 9 (47.4) 4 (19.0) 13 (32.5)
Weight, mean (SD), kg 79.9 (17.2) 92.1 (20.0) 86.1 (19.5)
Post-transplant, median (IQR), months 92 (37–185) 23 (15.5–51.0) 43.0 (20.5–131.7)
Transplant type
Kidney 18 (94.7) 21 (100.0) 39 (97.5)
Liver 1 (5.3) 0 (0.0) 1 (2.5)
Allograft function at randomization
Creatinine, mean (SD), lmol/L 120.1 (42.4) 122.2 (45.3) 121.2 (43.4)
eGFR, mean (SD), mL/min/1.73 m2 56.7 (23.9) 61.7 (24.5) 59.3 (24.0)
Year of transplant

Downloaded from https://academic.oup.com/ndt/article/35/6/1060/5855235 by guest on 06 November 2023


1980–89 1 (5.3) 0 (0.0) 1 (2.5)
1990–99 5 (26.3) 1 (4.8) 6 (15.0)
2000–09 8 (42.1) 4 (19.0) 12 (30.0)
2010þ 5 (26.3) 16 (76.2) 21 (52.5)
Hospital
Sir Charles Gairdner Hospital 10 (52.6) 17 (81.0) 27 (67.5)
RAH 9 (47.4) 4 (19.0) 13 (32.5)
Prior PPI 7 (36.8) 12 (57.1) 19 (47.5)
CNI type (total daily dose)
Cyclosporin 8 (42.1) 6 (28.6) 14 (35.0)
Dose, median (IQR), mg 150 (112–200) 200 (150–200) 171.4 (42.6)
Tacrolimus 11 (57.9) 15 (71.4) 26 (65.0)
Dose, median (IQR), mg 3 (2–4) 4 (2–7) 4.2 (3.3)
MPA (total daily dose)
MMF, median (IQR), mg 1500 (1000–2000) – –
EC-MPS, median (IQR), mg – 1440 (1080–1440) –
Prednisolone (total daily dose)
Dose, median (IQR), mg 5 (0–5) 5 (5–6) 5.7 (1.3)
Randomized drug
First drug placebo 9 (47.4) 11 (52.3) 20 (50.0)
First drug pantoprazole 10 (52.6) 10 (47.7) 20 (50.0)
Data are expressed as n (%), mean and SD or as median (IQR).

Of the 40 recipients, 19 (47.5%) and 21 (52.5%) were main- and Cmax and dose-normalized MPA-AUC12 h and Cmax were
tained on MMF and EC-MPS, respectively. Six (31.6%) recipi- significantly lower with pantoprazole compared with placebo.
ents were maintained on 2 g daily of MMF, and 11 (52.4%) There were no significant changes for Tmax or for all parameters
recipients were maintained on 1440 mg daily of EC-MPS at time for MPA-G with pantoprazole or placebo in both groups. For
of randomization. Recipients maintained on EC-MPS were recipients maintained on EC-MPS, MPA-AUC12 h and dose-
younger and were more likely to be recent transplant recipients normalized MPA-AUC12 h were significantly increased by
compared with those on MMF. Mean eGFR was 57 mL/min/ 25% with pantoprazole compared with placebo. There were
1.73 m2 for recipients maintained on MMF compared with no significant changes for Cmax, Tmax or for all parameters for
62 mL/min/1.73 m2 for recipients maintained on EC-MPS. Of MPA-G. The concentration–time profiles for MPA of recipients
the 19 recipients who were prescribed PPI prior to enrolment, maintained on MMF or EC-MPS 6 pantoprazole/placebo are
prior clinical symptom of gastro-oesophageal reflux disease was shown in Figure 2A and B, respectively.
the main indication for treatment. However, the ascertainment Table 3 shows the proportion of recipients maintained on
of the diagnosis or results of prior gastroscopy were not col- MMF or EC-MPS with MPA-AUC12 h of pre-specified thresh-
lected. Of recipients maintained on MMF, 58% were prescribed olds. Almost 90% of recipients maintained on MMF had MPA-
tacrolimus, whereas 71% of those on EC-MPS were prescribed AUC12 h of >30 mg*h/L, reducing to 68% with pantoprazole.
tacrolimus. The median doses of CNI, MMF, EC-MPS and pred- For recipients maintained on EC-MPS, 76% had MPA-AUC12 h
nisolone for MMF and EC-MPS groups are shown in Table 1. of >30 mg*h/L, increasing to 81% with pantoprazole.

Pharmacokinetics of MPA and MPA-G Cyclosporin and tacrolimus: pharmacokinetics of MPA


Table 2 shows the geometric means and 95% CI of MPA and and MPA-G
MPA-G concentrations for recipients maintained on MMF and Table 4 shows the geometric means and 95% CI of MPA-
EC-MPS, co-administered with placebo compared with panto- AUC12 h and Cmax concentrations for recipients maintained on
prazole. For recipients maintained on MMF, MPA-AUC12 h MMF and EC-MPS 6 placebo/pantoprazole and stratified by

1064 A. Sunderland et al.


Table 2. Pharmacokinetic parameters of MPA and MPA-G with concomitant placebo versus pantoprazole in kidney and liver transplant recipients
maintained on MMF or EC-MPS

Pharmacokinetic parameters (MMF and EC-MPS) Intervention Geometric means (95% CI) P-value
MMF: MPA
AUC12 h, mg*h/L Placebo 53.9 (44.0–65.9) 0.004
Pantoprazole 43.6 (35.6–53.4)
Dose-normalized AUC12 h, mg*h/La Placebo 76.0 (59.1–97.8) 0.005
Pantoprazole 61.6 (47.0–80.7)
Cmax, mg/L Placebo 16.1 (12.6–20.6) 0.002
Pantoprazole 10.7 (8.5–13.6)
Dose-normalized Cmax, mg/La Placebo 22.8 (18.3–28.3) 0.002
Pantoprazole 15.2 (11.6–19.8)
Tmax, h Placebo 1.0 (0.5–2.7) 0.422
Pantoprazole 1.2 (0.8–1.6)
MMF: MPA-G

Downloaded from https://academic.oup.com/ndt/article/35/6/1060/5855235 by guest on 06 November 2023


AUC12 h, mg*h/L Placebo 845.2 (678.2–1053.2) 0.856
Pantoprazole 825.8 (658.4–1035.8)
Dose-normalized AUC12 h, mg*h/La Placebo 1193.9 (955.7–1491.4) 0.747
Pantoprazole 1166.5 (920.2–1479.9)
Cmax, mg/L Placebo 99.5 (83.1–119.2) 0.573
Pantoprazole 94.4 (77.8–114.6)
Dose-normalized Cmax, mg/La Placebo 140.6 (119.0–166.1) 0.629
Pantoprazole 133.4 (108.7–163.7)
Tmax, h Placebo 2.2 (1.4–3.3) 0.639
Pantoprazole 2.3 (1.6–3.3)
EC-MPS: MPA
AUC12 h, mg*h/L Placebo 36.1 (26.5–49.2) 0.016
Pantoprazole 45.9 (35.5–59.3)
Dose-normalized AUC12 h (mg*h/L)b Placebo 41.3 (29.7–57.5) 0.023
Pantoprazole 52.6 (39.9–69.3)
Cmax, mg/L Placebo 8.5 (5.5–13.2) 0.211
Pantoprazole 11.9 (8.4–16.9)
Dose-normalized Cmax, mg/Lb Placebo 9.8 (6.2–15.4) 0.217
Pantoprazole 13.7 (9.5–19.6)
Tmax, h Placebo 3.1 (2.4–4.1) 0.577
Pantoprazole 3.9 (2.7–5.4)
EC-MPS: MPA-G
AUC12 h, mg*h/L Placebo 714.2 (559.2–912.1) 0.274
Pantoprazole 744.6 (571.4–970.4)
Dose-normalized AUC12 h, mg*h/Lb Placebo 818.0 (630.1–1062.0) 0.339
Pantoprazole 852.9 (655.8–1109.3)
Cmax, mg/L Placebo 84.5 (68.6–104.3) 0.159
Pantoprazole 87.6 (68.8–111.7)
Dose-normalized Cmax, mg/Lb Placebo 96.8 (76.8–121.9) 0.192
Pantoprazole 100.4 (78.6–128.2)
Tmax, h Placebo 2.6 (1.8–3.7) 0.180
Pantoprazole 4.3 (3.3–5.7)
Data are expressed as geometric means and 95% CIs, with Wilcoxon signed-rank test used to test the difference between placebo versus pantoprazole for each pharmacoki-
netic parameter. AUC12 h, AUC >12 h, Cmax, peak concentration; Tmax, time to peak concentration.
a
Dose-normalized to 2 g daily (in two equally divided doses) of MMF.
b
Dose-normalized to 1440 mg daily (in two equally divided doses) of EC-MPA.

CNI type. For recipients maintained on MMF and cyclosporin, MMF or EC-MPS and either tacrolimus or cyclosporin 6 panto-
MPA-AUC12 h and Cmax and dose-normalized MPA-AUC12 h prazole. Figure 3A and B shows the respective boxplots of MPA-
and Cmax were reduced by at least 20 and 30%, respectively, with AUC12 h and Cmax of recipients maintained on MMF and EC-
pantoprazole compared with placebo. For recipients maintained MPS, with placebo or pantoprazole and stratified by CNI type.
on MMF and tacrolimus, MPA-AUC12 h and Cmax and dose-
normalized MPA-AUC12 h and Cmax were significantly reduced Adverse events
by at least 15 and 30%, respectively, with pantoprazole. For There were no serious adverse events during the study period,
recipients maintained on EC-MPS and either cyclosporin or with no acute rejection, study drug discontinuation or modifica-
tacrolimus, there were no significant changes in MPA-AUC12 h tion of the immunosuppressive medications. A greater propor-
and Cmax with and without pantoprazole. There were no signifi- tion of recipients maintained on MMF experienced adverse
cant differences in MPA-G12 h for recipients maintained on events (with placebo and pantoprazole) compared with those

Effect of pantoprazole on MPA exposure 1065


A MMF + placebo/pantoprazole B EC–MPS + placebo/pantoprazole
18 8
Placebo: geometric mean and 95% CI Placebo: geometric mean and 95% CI
16 Pantoprazole: geometric mean and 95% CI Pantoprazole: geometric mean and 95% CI
7
14
Plasma MPA (mg/L)

Plasma MPA (mg/L)


12
5
10
4
8
3
6
4 2

2 1

0 0
0 0.5 1 1.5 2 3 4 5 6 8 10 12 0 0.5 1 1.5 2 3 4 5 6 8 10 12

Downloaded from https://academic.oup.com/ndt/article/35/6/1060/5855235 by guest on 06 November 2023


Time (h) Time (h)

FIGURE 2: Plasma MPA concentration–time profiles (geometric means and 95% CI) of recipients maintained on MMF (A) and EC-MPS
R
[MyforticV, (B)]. Co-administration of pantoprazole significantly reduced MPA exposure in recipients maintained on MMF, whereas the MPA
exposure was significantly increased in recipients maintained on EC-MPS.

Table 3. MPA-AUC thresholds in kidney and liver transplant recipients maintained on mycophenolate or EC-MPS with and without pantoprazole

MPA-AUC12 h thresholds, mg*h/L MMF (n ¼ 19) MMF þ pantoprazole (n ¼ 19) EC-MPS (n ¼ 21) EC-MPS þ pantoprazole (n ¼ 21)
>30 17 (89.5) 13 (68.4) 16 (76.2) 17 (81.0)
>40 15 (78.9) 10 (52.6) 10 (47.6) 14 (66.7)
>50 11 (57.9) 8 (42.1) 5 (23.8) 10 (47.6)
>60 10 (52.6) 7 (36.8) 4 (19.0) 9 (42.9)
Data are expressed as n (%).

Table 4. Pharmacokinetic parameters of MPA with concomitant placebo versus pantoprazole in kidney and liver transplant recipients maintained
on MMF or EC-MPS, stratified by calcineurin-inhibitor type (cyclosporin and tacrolimus)

Pharmacokinetic parameters (MMF and EC-MPS) Intervention Geometric means (95% CI)

Cyclosporin Tacrolimus
MMF: MPA
AUC12 h, mg*h/L Placebo 48.1 (34.8–66.5) 58.5 (43.5–78.6)
Pantoprazole 36.9 (26.0–52.4) 49.2 (37.6–64.3)*
Dose-normalized AUC12 h, mg*h/La Placebo 59.3 (41.2–85.3) 91.1 (64.2–129.4)
Pantoprazole 45.5 (28.1–73.7) 76.7 (56.5–104.3) *
Cmax, mg/L Placebo 16.5 (11.5–23.7) 15.9 (10.7–23.4)
Pantoprazole 11.1 (6.8–17.9) 10.5 (7.8–14.2)**
Dose-normalized Cmax, mg/La Placebo 20.4 (14.1–29.5) 24.7 (18.2–33.6)
Pantoprazole 13.6 (7.2–25.9) 16.4 (13.1–20.5)**
EC-MPS: MPA
AUC12 h, mg*h/L Placebo 19.5 (11.3–33.8) 46.2 (33.5–63.6)
Pantoprazole 30.9 (18.6–51.4) 53.8 (40.2–71.8)
Dose-normalized AUC12 h, mg*h/Lb Placebo 20.5 (11.5–36.3) 54.8 (39.5–75.9)
Pantoprazole 32.4 (17.8–58.9) 63.8 (47.8–85.1)
Cmax, mg/L Placebo 3.5 (1.4–8.7) 12.2 (8.0–18.5)
Pantoprazole 9.0 (4.2–19.3) 13.4 (8.7–20.5)
Dose-normalized Cmax, mg/Lb Placebo 3.7 (1.5–9.1) 14.4 (9.4–22.2)
Pantoprazole 9.4 (4.2–21.2) 15.9 (10.4–24.3)
Data are expressed as geometric means and 95% CIs, with Wilcoxon signed-rank test used to test the difference between placebo versus pantoprazole for each pharmacoki-
netic parameter.
a
Dose-normalized to 2 g daily (in two equally divided doses) of MMF.
b
Dose-normalized to 1440 mg daily (in two equally divided doses) of EC-MPA.
*P < 0.05,
**P < 0.01.

1066 A. Sunderland et al.


A MPA–AUC0-12h: MMF vs. EC–MPS B MPA–Cmax: MMF vs. EC–MPS
(+ pantoprazole/placebo by CNI type) (+ pantoprazole/placebo by CNI type)
MPA AUC-12h (mg*h/L) 150 50

MPA Cmax (mg/L)


40
100
30

20
50
10

0 0
MMF EC–MPS MMF EC–MPS MMF EC–MPS MMF EC–MPS
Tacrolimus Tacrolimus Tacrolimus Cyclosporin
MPA AUC-12h (placebo) MPA AUC-12h (PPI) MPA Cmax (placebo) MPA Cmax (PPI)

Downloaded from https://academic.oup.com/ndt/article/35/6/1060/5855235 by guest on 06 November 2023


FIGURE 3: Boxplots [showing the median, IQR, minimum and maximum and outliers (represented by solid circle)] of MPA AUC >12 h
R
[MPA-AUC12 h; (A)] and Cmax (B) for recipients maintained on either MMF or EC-MPS (MyforticV), stratified by CNI types (cyclosporin
and tacrolimus), with co-administration with placebo (blue bars) or pantoprazole (red bars).

Table 5. Adverse events of the study population

Adverse events of study cohort MMF (n ¼ 19) EC-MPS (n ¼ 21)


Adverse events Placebo Pantoprazole Placebo Pantoprazole
Any adverse events 8 (42.1) 7 (36.8) 4 (19.0) 2 (9.5)
Gastrointestinal 4 (21.1) 3 (15.6) 2 (9.5) 1 (4.8)
Reflux 2 (10.5) 2 (10.5) 1 (4.8) 1 (4.8)
Diarrhoea 1 (5.3) 1 (5.3) 0 (0.0) 0 (0.0)
Abdominal pain 1 (5.3) 1 (5.3) 0 (0.0) 1 (4.8)
Nausea/vomiting 1 (5.3) 1 (5.3) 1 (4.8) 0 (0.0)
Infection 2 (10.5) 3 (15.6) 0 (0.0) 0 (0.0)
Respiratory 1 0 0 0
Urinary tract 0 2 0 0
Others 1 0 0 0
Hospitalization 0 (0.0) 1 (5.3)b 0 (0.0) 0 (0.0)
Miscellaneous 2 (10.5) 1 (5.3) 3 (14.3) 0 (0.0)
Headache 1 1 1 0
Skin rash 1 0 0 0
Others 0 0 2 0
Haematology/biochemistrya
Creatinine, lmol/L 129 (80–154) 127 (80–161) 121 (91–138) 114 (85–149)
eGFR, mL/min/1.73 m2 50 (38–85) 49 (35–71) 56 (47–75) 54 (46–87)
White cell count (109/L) 6 (6–8) 4 (4–7) 7 (6–8) 7 (6–9)
Haemoglobin, g/L 130 (112–140) 128 (115–141) 138 (126–152) 137 (123–152)
Albumin, g/L 39 (38–42) 40 (38–42) 40 (38–43) 41 (38–42)
Allograft outcomes Placebo Pantoprazole Placebo Pantoprazole
Acute rejection 0 (0.0) 0 (0.0) 0 (0.0) 0 (0.0)
Allograft failure 0 (0.0) 0 (0.0) 0 (0.0) 0 (0.0)
Death 0 (0.0) 0 (0.0) 0 (0.0) 0 (0.0)
Data are expressed as n (%).
a
Expressed as median (IQR).
b
Hospitalization secondary to urinary tract infection and dehydration.

maintained on EC-MPS, and the majority of adverse events were on placebo and pantoprazole, regardless of whether they were
mild non-specific symptoms (headache and/or mild gastro- maintained on MMF or EC-MPS. There were no significant dif-
intestinal symptoms). The proportion of recipients who had ex- ferences in serum haemoglobin, albumin or creatinine (and
perienced reflux symptoms was similar for recipients maintained eGFR) between the two pharmacokinetic timepoints in either

Effect of pantoprazole on MPA exposure 1067


group of recipients maintained on MMF or EC-MPS. Table 5 resulted in a significant earlier Tmax when co-administered with
shows the adverse events for recipients maintained on MMF and EC-MPS, but there was no effect on MPA-AUC12 h or Cmax
EC-MPS, stratified by placebo and pantoprazole. [19]. Given the small number of recipients and the large inter-
individual pharmacokinetic variability, the true effect of PPI on
Sensitivity analysis MPA exposure remains uncertain. Similarly, PPI-induced re-
In the sensitivity analysis excluding the one liver transplant duction in MPA exposure in 22 heart transplant recipients
recipient (of recipients maintained on MMF), the geometric maintained on MMF has been reported [8], but a similar effect
means (95% CI) of each pharmacokinetic parameter in the was not observed for heart or lung recipients maintained on
presence of placebo and pantoprazole had remained similar EC-MPS (n ¼ 21) [9]. Similarly, in a retrospective study of 61
(Supplementary data, Table S1). When the study population kidney transplant recipients, lansoprazole reduced the MPA ex-
was stratified according to eGFR thresholds of 60 and posure in recipients maintained on MMF and tacrolimus, but
>60 mL/min/1.73 m2, the estimates for actual and dose- this work left open the possibility that the drug–drug interac-
normalized MPA and MPA-G AUC12 h and Cmax were similar tion may be dependent on the PPI type and cytochrome P450
to the estimates from the main analysis for recipients main- and efflux transporter genetic polymorphisms (e.g. CYP2C19

Downloaded from https://academic.oup.com/ndt/article/35/6/1060/5855235 by guest on 06 November 2023


tained on MMF and EC-MPS, although statistically significant and ABCB1 polymorphisms) [20]. Nevertheless, these studies
difference in the change between MPA-AUC12 h and/or Cmax in were not randomized controlled trials and therefore the clinical
the presence of placebo versus pantoprazole was evident only significance of the study findings must be interpreted with cau-
for recipients with eGFR of 60 mL/min/1.73 m2. In recipients tion. In our study, co-administration of pantoprazole with
with eGFR of >60 mL/min/1.73 m2, the proportional change in MMF significantly reduced the total MPA exposure (MPA-
MPA-AUC12 h and Cmax was not statistically significant but was AUC12 h and Cmax), which was not observed in recipients main-
similar to recipients with eGFR of 60 mL/min/1.73 m2 tained on EC-MPS. However, pantoprazole had no effect on
(Supplementary data, Table S2). MPA Tmax or the pharmacokinetic profile of MPA-G (which
should reflect the total amount of absorbed MPA) in recipients
maintained on MMF or EC-MPS, which may reflect the large
DISCUSSION inter-patient variability, potential differences polymorphisms of
This randomized, double-blind placebo-controlled cross-over uridine 50 -diphospho-glucuronosyltransferase genes (enzymes
study showed an important interaction between pantoprazole involved in the metabolism of MPA), the pre-specified sam-
and MPA formulations in stable kidney and liver transplant pling points to calculate the pharmacokinetic profile may have
recipients maintained on CNI-based immunosuppression. missed the true Cmax, and the potential variability in the renal
Consistent with the known increase in gastric pH with the use excretion and the entero-hepatic recycling of MPA-G [21, 22].
of PPI, co-administration of pantoprazole reduced the bioavail- The finding that pantoprazole significantly increased the MPA-
ability of MPA in patients maintained on MMF but had an en- AUC12 h and dose-normalized AUC12 h following EC-MPS ad-
hanced effect on the MPA bioavailability in those maintained ministration suggests that absorption of EC-MPS occurred ear-
on EC-MPS. lier in the presence of pantoprazole even though the Cmax
The pharmacokinetic profiles of the two MPA formulations attained was not dissimilar to the absence of pantoprazole.
of MMF and EC-MPS are dissimilar, with the absorption re- Nevertheless, it has been shown that the MPA pharmacokinetic
quiring an acidic and neutral pH environment, respectively, be- profile for EC-MPS exhibits a greater intra-patient variability
fore making active MPA available. As such, MPA when compared with MMF formulation, which may have con-
concentrations peak relatively earlier, within 1–2 h, following tributed to differences between our study and previous reports
MMF administration. In contrast, peak MPA concentrations [23]. Differences in the study design, sample size, population
occur between 2 and 4 h after EC-MPS [15–17]. Consequently, characteristics and concomitant CNI maintenance may also
it has been shown that PPI, by suppressing gastric acid secretion have contributed to the dissimilar findings between our study
will interfere with the absorption and reduce the total MPA ex- and the cross-over study from Germany [19].
posure following MMF but not EC-MPS. In two randomized The systemic exposure of MMF and EC-MPS has been
cross-over studies (not placebo-controlled) of healthy volun- shown to be generally equivalent after equimolar dosing (i.e.
teers, concomitant PPI treatment (n ¼ 12 omeprazole 20 mg MPA-AUC0–1 following 720 mg EC-MPS is equivalent to 1 g
twice daily and n ¼ 12 pantoprazole 40 mg twice daily) signifi- of MMF: 66.5 lg*h/mL versus 63.7 lg*h/mL, respectively) [16,
cantly reduced the MPA and MPA-G AUC12 h and Cmax with 21]. Several randomized controlled trials and cohort studies
MMF but not with EC-MPS [10, 18]. In both studies, the volun- have shown that higher MPA exposure (MMF formulation)
teers were exposed only to a single daily dose of MMF (1 g) and may be associated with a lower risk of acute rejection, particu-
EC-MPS (720 mg). In a randomized 4  4 cross-over single- larly achieving MPA-AUC >30 mg*h/L in the first week post-
centre German study (2-week periods each of MMF, EC-MPS, transplant, but this association was no longer apparent beyond
EC-MPS/pantoprazole and MMF/pantoprazole) of 18 kidney this timepoint [4, 5, 24–27]. The clinical relevance of MPA con-
transplant recipients maintained on cyclosporin-based regimen, centration in the outcome of other solid organ transplants (in-
there was at least a respective 10 and 20% reduction in MPA- cluding liver and heart transplant recipients) remains unclear,
AUC12 h and Cmax when MMF was co-administered with pan- with most studies evaluating MPA trough levels rather than ex-
toprazole, but this was not statistically significant. Pantoprazole tended MPA profiles [28–32]. There have been no clinical

1068 A. Sunderland et al.


studies that have examined the association between MPA expo- concentrations by almost 20%, but the longer term clinical ef-
sure and allograft outcome in kidney transplant recipients fect of this interaction remains unknown. Even though the clin-
maintained on EC-MPS. In our study, a greater proportion of ical significance of MPA TDM in chronic kidney and liver
recipients maintained on MMF had achieved higher mean transplant recipients remains uncertain, clinicians should still
MPA-AUC12 h, despite a smaller proportion being maintained be cognizant of this important drug interaction when prescrib-
on the ‘maximal bio-equivalent dose’ of MMF (2 g/day, 32%) ing PPI with MMF or EC-MPS in the early post-transplant pe-
compared with EC-MPS (1440 mg/day, 52%). In the presence riod. Future studies designed to evaluate the allograft outcome
of pantoprazole, the proportion of recipients maintained on of recipients maintained on different MPA formulation with
MMF with MPA-AUC12 h >30 mg*h/L was reduced by 22% and without PPI are needed to ascertain the long-term clinical
compared with placebo, which may have potential clinical rele- relevance of this drug interaction.
vance in the early post-transplant period. For recipients main-
tained on EC-MPS, pantoprazole substantially increased the
SUPPLEMENTARY DATA
proportion of recipients achieving higher MPA-AUC12 h
thresholds, particularly >40 mg*h/L. Supplementary data are available at ndt online.

Downloaded from https://academic.oup.com/ndt/article/35/6/1060/5855235 by guest on 06 November 2023


Co-administration of different CNI types has been shown to
influence MPA exposure with the use of cyclosporin being associ- ACKNOWLEDGEMENTS
ated with up to 50% reduction in MPA exposure compared with
tacrolimus. This has been attributed to the differential effect of The authors are grateful for the unrestricted educational grant
R

the different CNIs on enterohepatic recycling [21, 33, 34]. In our provided by NovartisV for the completion of this trial; how-
study, the MPA-AUC12 h and dose-normalized AUC12 h were ever, the study design, conduct of the trial, interpretation of
higher for tacrolimus-treated patients compared with the data, analysis, writing of the paper and decision to submit
cyclosporin-treated patients, despite a higher median dose of the manuscript for publication were undertaken only by the
R

MMF and EC-MPS being prescribed for cyclosporin-treated authors, without involvement by NovartisV.
patients. The co-administration of pantoprazole reduced the
MPA-AUC12 h and Cmax for tacrolimus- and cyclosporin-treated FUNDING
patients, but this was not statistically significant (with wide CIs)
W.H.L. is supported by a Clinical Research Fellowship from
in the latter group, likely reflecting the small number of patients
the Raine Foundation (University of Western Australia and
maintained on cyclosporin. Similarly, when stratified by eGFR of
Health Department of Western Australia) and Jacquot
thresholds of 60 and >60 mL/min/1.73 m2, the results were
Research Foundation (Royal Australasian College of
similar although the proportional change in MPA-AUC12 h and
Physicians). G.W. is supported by a National Health and
Cmax was not statistically significant for recipients with entry
Medical Research Council Career Development Fellowship.
eGFR of >60 mL/min/1.73 m2, likely to reflect the smaller sample
E.O. is supported by a National Heart Foundation Future
size.
Leader Fellowship (Award ID: 102538).
There are a few limitations in this study. Our sample size cal-
culation was based on an effect size of 25% in AUC12 h, and
when combined with the variability in MPA pharmacokinetic AUTHORS’ CONTRIBUTIONS
profiles may suggest that our study was not sufficiently powered
W.H.L. and G.R. designed the study and recruited the partici-
for certain pharmacokinetic parameters, particularly when
pants. W.H.L. has full access to the trial data. W.H.L. and
stratified by CNI types. Nevertheless, this study remains the
E.O. participated in the analysis of the data. All authors
largest randomized cross-over trial conducted in stable kidney
participated in the interpretation of the data and writing of
and liver transplant recipients. It is possible that the 1 week
the article.
washout period for pantoprazole was inadequate, but given the
plasma half-life of pantoprazole is around 1 h [35], it is unlikely
that there was any residual carry-over effect of pantoprazole CONFLICT OF INTEREST STATEMENT
during the wash-out period. In addition, the dose of 40 mg daily N.B. has received speaking honoraria from Baxter, travel
of pantoprazole was chosen as the intervention to reflect regi- grants from Amgen and Roche, and unrestricted educational
mens most commonly used in clinical transplant practice in grants from Amgen, Roche and Baxter. W.H.L. has received
Australia, but the study findings may not be able to extrapolate speaking honoraria from Alexion, Astellas and Novartis, and
to other PPIs, which may have dissimilar half-lives and inhibi- unrestricted educational grants from Astellas and Novartis.
tory effects on gastric acid secretion [36]. Given, we had only
recruited one liver transplant recipient, our findings cannot be
extrapolated to this population. Furthermore, our study find- REFERENCES
ings may not be readily generalizable across kidney transplant 1. Ekberg H, Tedesco-Silva H, Demirbas A et al. Reduced exposure to calci-
recipients with poorer allograft function or in recipients with neurin inhibitors in renal transplantation. N Engl J Med 2007; 357:
2562–2575
long-term exposure to PPI. 2. Le Meur Y, Buchler M, Thierry A et al. Individualized mycophenolate mofe-
Despite the pharmacokinetic variability of MPA, the concur- til dosing based on drug exposure significantly improves patient outcomes
rent administration of PPI substantially reduced the MPA after renal transplantation. Am J Transplant 2007; 7: 2496–2503

Effect of pantoprazole on MPA exposure 1069


3. Knight SR, Morris PJ. Does the evidence support the use of mycophenolate 21. Shaw LM, Figurski M, Milone MC et al. Therapeutic drug monitoring of
mofetil therapeutic drug monitoring in clinical practice? A systematic re- mycophenolic acid. Clin J Am Sci Nephrol 2007; 2: 1062–1072
view. Transplantation 2008; 85: 1675–1685 22. Levesque E, Delage R, Benoit-Biancamano MO et al. The impact of
4. Gaston RS, Kaplan B, Shah T et al. Fixed- or controlled-dose mycophenolate UGT1A8, UGT1A9, and UGT2B7 genetic polymorphisms on the pharma-
mofetil with standard- or reduced-dose calcineurin inhibitors: the Opticept cokinetic profile of mycophenolic acid after a single oral dose in healthy vol-
trial. Am J Transplant 2009; 9: 1607–1619 unteers. Clin Pharmacol Ther 2007; 81: 392–400
5. van Gelder T, Silva HT, de Fijter JW et al. Comparing mycophenolate mofe- 23. Cattaneo D, Cortinovis M, Baldelli S et al. Pharmacokinetics of mycopheno-
til regimens for de novo renal transplant recipients: the fixed-dose concen- late sodium and comparison with the mofetil formulation in stable kidney
tration-controlled trial. Transplantation 2008; 86: 1043–1051 transplant recipients. Clin J Am Sci Nephrol 2007; 2: 1147–1155
6. Helderman JH, Goral S. Gastrointestinal complications of transplant immu- 24. Pawinski T, Durlik M, Szlaska I et al. Comparison of mycophenolic acid
nosuppression. J Am Soc Nephrol 2002; 13: 277–287 pharmacokinetic parameters in kidney transplant patients within the first 3
7. Kofler S, Deutsch MA, Bigdeli AK et al. Proton pump inhibitor co- months post-transplant. J Clin Pharm Ther 2006; 31: 27–34
medication reduces mycophenolate acid drug exposure in heart transplant 25. Sanchez Fructuoso AI, de la Higuera MA, Garcia-Ledesma P et al. Graft
recipients. J Heart Lung Transplant 2009; 28: 605–611 outcome and mycophenolic acid trough level monitoring in kidney trans-
8. Kofler S, Shvets N, Bigdeli AK et al. Proton pump inhibitors reduce myco- plantation. Transplant Proc 2009; 41: 2102–2103
phenolate exposure in heart transplant recipients-a prospective case- 26. Sanchez-Fructuoso AI, de la Higuera MA, Giorgi M et al. Inadequate myco-
controlled study. Am J Transplant 2009; 9: 1650–1656 phenolic acid exposure and acute rejection in kidney transplantation.

Downloaded from https://academic.oup.com/ndt/article/35/6/1060/5855235 by guest on 06 November 2023


9. Kofler S, Wolf C, Shvets N et al. The proton pump inhibitor pantoprazole Transplant Proc 2009; 41: 2104–2105
and its interaction with enteric-coated mycophenolate sodium in transplant 27. Gourishankar S, Houde I, Keown PA et al. The CLEAR study: a 5-day,
recipients. J Heart Lung Transplant 2011; 30: 565–571 3-g loading dose of mycophenolate mofetil versus standard 2-g
10. Rupprecht K, Schmidt C, Raspe A et al. Bioavailability of mycophenolate dosing in renal transplantation. Clin J Am Sci Nephrol 2010; 5:
mofetil and enteric-coated mycophenolate sodium is differentially affected 1282–1289
by pantoprazole in healthy volunteers. J Clin Pharmacol 2009; 49: 28. Saliba F, Rostaing L, Gugenheim J et al. Corticosteroid-sparing and optimi-
1196–1201 zation of mycophenolic acid exposure in liver transplant recipients receiving
11. Westley IS, Sallustio BC, Morris RG. Validation of a high-performance mycophenolate mofetil and tacrolimus: a randomized, multicenter study.
liquid chromatography method for the measurement of mycophenolic Transplantation 2016; 100: 1705–1713
acid and its glucuronide metabolites in plasma. Clin Biochem 2005; 38: 29. Meiser BM, Pfeiffer M, Schmidt D et al. Combination therapy with tacroli-
824–829 mus and mycophenolate mofetil following cardiac transplantation: impor-
12. Hale MD, Nicholls AJ, Bullingham RE et al. The pharmacokinetic- tance of mycophenolic acid therapeutic drug monitoring. J Heart Lung
pharmacodynamic relationship for mycophenolate mofetil in renal trans- Transplant 1999; 18: 143–149
plantation. Clin Pharmacol Ther 1998; 64: 672–683 30. Yamani MH, Starling RC, Goormastic M et al. The impact of routine myco-
13. van Gelder T, Hilbrands LB, Vanrenterghem Y et al. A randomized double- phenolate mofetil drug monitoring on the treatment of cardiac allograft re-
blind, multicenter plasma concentration controlled study of the safety and jection. Transplantation 2000; 69: 2326–2330
efficacy of oral mycophenolate mofetil for the prevention of acute rejection 31. Tredger JM, Brown NW, Adams J et al. Monitoring mycophenolate in liver
after kidney transplantation. Transplantation 1999; 68: 261–266 transplant recipients: toward a therapeutic range. Liver Transpl 2004; 10:
14. van Gelder T, Le Meur Y, Shaw LM et al. Therapeutic drug monitoring of 492–502
mycophenolate mofetil in transplantation. Ther Drug Monit 2006; 28: 32. Kuypers DR, Le Meur Y, Cantarovich M et al. Consensus report on thera-
145–154 peutic drug monitoring of mycophenolic acid in solid organ transplanta-
15. Bullingham RE, Nicholls AJ, Kamm BR. Clinical pharmacokinetics of tion. Clin J Am Sci Nephrol 2010; 5: 341–358
mycophenolate mofetil. Clin Pharmacokinet 1998; 34: 429–455 33. van Gelder T, Klupp J, Barten MJ et al. Comparison of the effects of tacroli-
16. Arns W, Breuer S, Choudhury S et al. Enteric-coated mycophenolate so- mus and cyclosporine on the pharmacokinetics of mycophenolic acid. Ther
dium delivers bioequivalent MPA exposure compared with mycophenolate Drug Monit 2001; 23: 119–128
mofetil. Clin Transplant 2005; 19: 199–206 34. Grinyo JM, Ekberg H, Mamelok RD et al. The pharmacokinetics of myco-
17. Lidgate D, Brandl M, Holper M et al. Influence of ferrous sulfate on the sol- phenolate mofetil in renal transplant recipients receiving standard-dose or
ubility, partition coefficient, and stability of mycophenolic acid and the ester low-dose cyclosporine, low-dose tacrolimus or low-dose sirolimus: the
mycophenolate mofetil. Drug Dev Ind Pharm 2002; 28: 1275–1283 Symphony pharmacokinetic substudy. Nephrol Dial Transplant 2009; 24:
18. Kees MG, Steinke T, Moritz S et al. Omeprazole impairs the absorption of 2269–2276
mycophenolate mofetil but not of enteric-coated mycophenolate sodium in 35. Huber R, Hartmann M, Bliesath H et al. Pharmacokinetics of pantoprazole
healthy volunteers. J Clin Pharmacol 2012; 52: 1265–1272 in man. Int J Clin Pharmacol Ther 1996; 34: 185–194
19. Rissling O, Glander P, Hambach P et al. No relevant pharmacokinetic inter- 36. Shin JM, Sachs G. Pharmacology of proton pump inhibitors. Curr
action between pantoprazole and mycophenolate in renal transplant patients: Gastroenterol Rep 2008; 10: 528–534
a randomized crossover study. Br J Clin Pharmacol 2015; 80: 1086–1096
20. Miura M, Satoh S, Inoue K et al. Influence of lansoprazole and rabeprazole Received: 30.1.2020; Editorial decision: 31.3.2020
on mycophenolic acid pharmacokinetics one year after renal transplanta-
tion. Ther Drug Monit 2008; 30: 46–51

1070 A. Sunderland et al.

You might also like