Lack of genetic association between OCT1, ABCB1, and UGT2B7 variants and morphine pharmacokinetics

Lack of genetic association between OCT1, ABCB1, and UGT2B7 variants and morphine pharmacokinetics

Accepted Manuscript Lack of genetic association between OCT1, ABCB1, and UGT2B7 variants and morphine pharmacokinetics LM Nielsen, E Sverrisdóttir, T...

544KB Sizes 0 Downloads 95 Views

Accepted Manuscript Lack of genetic association between OCT1, ABCB1, and UGT2B7 variants and morphine pharmacokinetics

LM Nielsen, E Sverrisdóttir, TB Stage, S Feddersen, K Brøsen, LL Christrup, AM Drewes, AE Olesen PII: DOI: Reference:

S0928-0987(16)30580-2 doi: 10.1016/j.ejps.2016.12.039 PHASCI 3856

To appear in:

European Journal of Pharmaceutical Sciences

Received date: Revised date: Accepted date:

21 August 2016 20 November 2016 31 December 2016

Please cite this article as: LM Nielsen, E Sverrisdóttir, TB Stage, S Feddersen, K Brøsen, LL Christrup, AM Drewes, AE Olesen , Lack of genetic association between OCT1, ABCB1, and UGT2B7 variants and morphine pharmacokinetics. The address for the corresponding author was captured as affiliation for all authors. Please check if appropriate. Phasci(2017), doi: 10.1016/j.ejps.2016.12.039

This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

ACCEPTED MANUSCRIPT

Lack of genetic association between OCT1, ABCB1, and UGT2B7 variants and morphine pharmacokinetics Nielsen LM*1,2, Sverrisdóttir E*2, Stage TB3, Feddersen S4, Brøsen K3, Christrup LL2, Drewes

PT

AM1,5 & Olesen AE1,2,5

RI

*These authors contributed equally to this work

SC

1. Mech-Sense, Department of Gastroenterology and Hepatology, Aalborg University Hospital, Aalborg, Denmark

NU

2. Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences,

MA

University of Copenhagen, Copenhagen, Denmark

3. Clinical Pharmacology, Department of Public Health, University of Southern Denmark,

D

Odense, Denmark

Odense, Denmark

PT E

4. Department of Clinical Biochemistry and Pharmacology, Odense University Hospital,

CE

5. Department of Clinical Medicine, Aalborg University, Aalborg, Denmark

AC

Running head: Lack of association between gene polymorphisms and morphine PK

1

ACCEPTED MANUSCRIPT

Corresponding author:

Anne Estrup Olesen, MSc (pharm), PhD Mech-Sense, Department of Gastroenterology & Hepatology Medicinerhuset, Mølleparkvej 4, 3rd floor

PT

Aalborg University Hospital

RI

DK-9000 Aalborg, Denmark

SC

Telephone: +45 9766 0535

AC

CE

PT E

D

MA

NU

E-mail: [email protected]

2

ACCEPTED MANUSCRIPT

ABSTRACT Aim A high inter-individual variation in the pharmacokinetics and pharmacodynamics of morphine has been observed. Genetic polymorphisms in genes encoding the organic cation transporter isoform 1

PT

(OCT1), the efflux transporter p-glycoprotein (ABCB1), and the UDP-glucuronosyltransferase-2B7 (UGT2B7) may influence morphine pharmacokinetics and thus, also pharmacodynamics. The aim

RI

of this study was to evaluate the association between OCT1, ABCB1, and UGT2B7 variants, and

SC

morphine pharmacokinetics and -dynamics in healthy volunteers. Methods

NU

Pharmacokinetic and pharmacodynamic data were collected from a double-blinded, randomized,

MA

crossover trial in 37 healthy subjects. Pharmacokinetic data were analyzed in NONMEM®, and the time-concentration relationship of morphine, morphine-3-glucuronide, and morphine-6-glucuronide

D

was parameterized as the transit compartment rate constant (ktr), clearance (CL), and volume of

PT E

distribution (VD). The area under the plasma concentration-time curve (AUC0-150min) and the maximum plasma concentration (Cmax) were also calculated. Pharmacodynamic data were measured as pain tolerance thresholds to mechanical stimulation of the rectum and muscle, as well as tonic

CE

cold pain stimulation (“the cold pressor test” where hand was immersed in cold water). Six different

AC

single nucleotide polymorphisms in three different genes (OCT1 (n=22), ABCB1 (n=37), and UGT2B (n=22)) were examined. Results

Neither AUC0-150min, ktr, CL, nor VD were associated with genetic variants in OCT1, ABCB1, and UGT2B7 (all P>0.05). Similarly, the antinociceptive effects of morphine on rectal, muscle, and cold pressor tests were not associated with these genetic variants (all P>0.05).

3

ACCEPTED MANUSCRIPT

Conclusions In this experimental study in healthy volunteers, we found no association between different genotypes of OCT1, ABCB1, and UGT2B7, and morphine pharmacokinetics and pharmacodynamics. Nonetheless, due to methodological limitations we cannot exclude that

RI

PT

associations exist.

SC

Keywords: Morphine, Interindividual variability, Pharmacokinetics, Pharmacogenetics, Healthy

AC

CE

PT E

D

MA

NU

volunteers

4

ACCEPTED MANUSCRIPT

1. INTRODUCTION Morphine is a widely prescribed opioid for the treatment of severe acute and chronic pain. However, patients respond differently to morphine and this inter-individual variability is still a major challenge in the clinical use of morphine [1]. About 10-30 % of patients treated with

PT

morphine do not have adequate treatment outcome either because of inadequate analgesia or intolerable side effects [2]. Some of the inter-individual variability in the pharmacokinetic fate of

RI

morphine is caused by age, organ function, gender and co-morbidity [3]. However, gene variants

SC

relevant to opioid absorption, distribution, metabolism and excretion may also influence the

NU

pharmacokinetics and thus, the pharmacodynamics of morphine [3].

MA

Organic cation transporter 1 (OCT1) is expressed in the human hepatocytes and is involved in the distribution of organic cations, such as morphine [4]. OCT1 is highly genetically variable and five

D

common reduced-function OCT1 single nucleotide polymorphisms (SNPs) are known in

PT E

Caucasians [5, 6]. Around 10 % of Caucasians have substantially reduced or absent OCT1 activity and the OCT1 reduced-function allele limits the uptake and activity of drugs metabolized in the

CE

human liver [7–9]. Morphine has previously been shown to be an OCT1 substrate [10], and thus, it

AC

could be speculated that OCT1 reduced-function allele may affect morphine pharmacokinetics [11].

The efflux transporter P-glycoprotein, encoded by the ATP-binding cassette B1 (ABCB1) gene, have a major impact on the brain distribution of opioids such as morphine and its metabolites, morphine-3-glucuronide (M3G) and morphine-6-glucuronide (M6G) [12, 13]. More than 50 SNPs and 3 insertion/deletion polymorphisms in ABCB1 have been suggested to affect the efflux activity of the transporter and thus the distribution of many drugs [14]. One polymorphism of interest is the SNP C3435T (rs1045642), that is associated with altered expression and function of P-glycoprotein,

5

ACCEPTED MANUSCRIPT

and consequently, this has led to altered pharmacokinetics and pharmacodynamics of different analgesics including morphine although the mechanisms are still unclear [15, 16]. The variant Tallele is found to be linked with lower P-glycoprotein expression and/or activity in the duodenum [17], natural killer cells [18], and renal parenchyma [19]. In animals, the lack of functional P-

PT

glycoprotein has been shown to have an important impact on analgesia after administration of opioids including morphine [20, 21]. A downregulation of the P-glycoprotein expression reduced

RI

the brain-to-blood transport of morphine and therefore, lowered P-glycoprotein expression

SC

significantly enhanced systemic morphine analgesia [21]. These findings indicate that genetic variation in P-glycoprotein may influence morphine absorption, distribution metabolism, and

NU

elimination due to it is located in the intestine, blood-brain-barrier, liver, and kidneys [22], and

MA

thereby, alter morphine pharmacokinetics and -dynamics.

D

Morphine is primarily metabolized by UGT2B7 to 90 % M3G and 10 % M6G [1, 23]. In one study

PT E

UGT2B7 variants have been shown to result in differences in glucuronidation activity [24]. However, the reported effect of UGT2B7 variants on glucuronidation activity is still inconsistent across studies. The UGT2B7 372 A>G (rs28365063) polymorphism has been reported to increase

CE

glucuronidation activity for morphine [25]. Additionally, the UGT2B7 -900 G>A (rs7438135)

AC

polymorphism has been associated with altered morphine pharmacokinetic in preterm infants and in young adults with sickle cell disease [26, 27].

Based on results from studies conducted so far, the influence of polymorphisms in OCT1, ABCB1, and UGT2B7 on clinical response seen after morphine administration is unclear. Investigation on the association between polymorphisms in these genes and morphine response in human

6

ACCEPTED MANUSCRIPT

experimental pain models might contribute with important knowledge, since confounders are limited compared to studies in patients [28, 29].

We hypothesized that genetic variation in OCT1, ABCB1, and UGT2B7 might influence the

PT

morphine pharmacokinetics and thereby, also influence morphine pharmacodynamics. This was investigated in a multi-modal, multi-tissue experimental pain setup in 40 healthy volunteers. The

RI

overall purpose of the study was 1) to evaluate associations between polymorphisms in OCT1,

SC

ABCB1 and UGT2B7, and the pharmacokinetics, measured as area under the plasma concentrationtime curve (AUC0-150min), the transit compartment rate constant (ktr), the clearance (CL), the volume

NU

of distribution (VD), and the maximum plasma concentration (Cmax) of morphine, M3G and M6G.

MA

And 2) to investigate if these polymorphisms were associated with morphine pharmacodynamics, measured as pain tolerance thresholds to rectal pressure stimulation, muscle pressure stimulation

AC

CE

PT E

D

and the cold pressor test (immersion of the hand into ice water).

7

ACCEPTED MANUSCRIPT

2. MATERIAL AND METHODS 2.1 Study procedure The original study was conducted in the period November 2010 to April 2012. Pharmacokinetic and pharmacodynamics data were collected from this double-blinded, randomized, crossover trial with a

PT

washout period of at least 1 week between visits. Forty healthy Caucasians were randomized into

RI

four blocks (10 participants in each block receiving the same order of the study medication) to

SC

receive either oral morphine or placebo on day 1 or day 2. The participants received 30 mg (2 mg/mL) of oral morphine or 15 mL placebo. The North Denmark Region Committee on Health

NU

Research Ethics (reference no. N-20100046) and Danish Medicines Agency (reference no. 26124319) approved the study. The trial was registered at ClinicalTrials.gov with identifier number

MA

NCT01245244. Pharmacodynamic results of placebo versus morphine and population pharmacokinetic-pharmacodynamic models for morphine are presented in separate published papers

D

[30, 31]. Furthermore, the genetic influence of mu opioid receptor (OPRM1), kappa opioid receptor

PT E

(OPRK1), delta opioid receptor (OPRD1), and catechol-O-methyltransferase (COMT) on the pain sensitivity has been published (submitted) prior to the current publication. Genotyping of the

CE

ABCB1 SNP rs1045642 was carried out after the study was completed.

AC

An add-on study was performed to investigate the association between polymorphisms in genes encoding the transporter (OCT1) and the drug-metabolizing enzyme (UGT2B7), and morphine pharmacokinetics. This study was approved by The North Denmark Region Committee on Health Research Ethics (reference no N-20140018), and was carried out in the Research Laboratory at Mech-Sense, Department of Gastroenterology and Hepatology, Aalborg University Hospital, Aalborg, Denmark. The study was conducted in the period September 2014 to April 2015 and consisted of one study day, where two blood samples were collected with the purpose of genotyping

8

ACCEPTED MANUSCRIPT

for genetic variants in above mentioned genes. A research biobank of the genetic blood samples was established. 2.2 Study participants All 40 participants from the original study were contacted again if possible. Inclusion criteria were:

PT

1) age between 20 and 65 years; 2) opioid naïve; 3) no known allergy to morphine; 4) no ongoing

RI

participation in other drug studies; 5) not pregnant (all included woman were on safe contraceptive medication during the study); 6) no previous addictive behavior; 7) no previous pain causing

SC

diseases or psychiatric disorders. Exclusion criteria were not relevant for this study. Participants

NU

gave their informed consent before included in the study.

MA

2.3 Blood sampling and quantitative analysis of morphine and its metabolites Blood samples of 6 mL were collected in K2EDTA tubes at fixed times before and after drug

D

administration (t = 0 (drug administration), 5, 10, 15, 30, 45, 60, 90, 120, and 150 min). Plasma was

PT E

separated and kept at -80oC until analysis. The concentrations of morphine, M3G and M6G were determined using ultra-high-performance liquid chromatography/mass spectrometry as previously

CE

described [31].

2.4 Pharmacokinetic variables and data analysis

AC

The population pharmacokinetic model is presented in our previously publication [31]. Briefly, individual morphine concentration-time data were fitted to pharmacokinetic models using nonlinear mixed effects modelling (NONMEM version 7.2; ICON Plc). Morphine pharmacokinetics was best described with a one-compartment model, which was parameterized in terms of clearance (CL, L/min) and volume of distribution (VD, L). The absorption of morphine after administration in oral solution was described with a two compartment transit absorption model with a transit compartment rate constant (ktr, min-1). Individual predicted parameter estimates of ktr, CL, and VD for morphine,

9

ACCEPTED MANUSCRIPT

M3G, and M6G were used in the data analysis in the current publication. The AUC0-150 min of morphine was calculated in R [32], based on individual predicted concentrations. Cmax values were the maximum individual predicted concentrations. Parameter estimates are listed as medians with 25th to 75th percentile in Table 2.

PT

2.5 Pharmacodynamic variables and data analysis

RI

Data were collected from the original study and details of the experimental pain tests have been described previously [30]. In this study experimental pain testing in the healthy volunteers included

SC

rectal pressure stimulation with a bag, muscle pressure stimulation at the left forearm and the cold

NU

pressor test to left hand. These three tests were selected in order to limit multiple testing and they showed the smallest variation between subjects. The experimental tests were used as a “control”

MA

measurement to see if an altered morphine pharmacokinetics affect morphine pharmacodynamics. All stimulations were interrupted when the participants reported pain tolerance threshold (PTT) on a

D

validated modified numerical rating scale from 0 to 10, where 5 = the pain detection threshold and 7

PT E

= PTT [33].

Percentage change from baseline ((T60 min-baseline)/baseline*100%) for morphine (Δmor) and

CE

placebo (Δpla) effects in: 1) tolerated volume (mL) at PTT level for rectal stimulation; 2) tolerated

AC

pressure (kPa) at PTT level for muscle stimulation; and 3) area under the time-pain level curve (ice AUC) for the cold pressor test. Morphine response was calculated as difference between morphine and placebo effects (Δmor – Δpla) and presented as percentage points (PP). 2.6 DNA extraction and genotyping DNA was extracted from an aliquot of venous blood with the QIAsymphony DSP DNA Midi kit (Qiagen, Copenhagen, Denmark) using the QIAsymphonyTM SP instrument (Qiagen, Copenhagen, Denmark). Selected SNPs in OCT1 (rs12208357) and UGT2B7 (rs7438135 and rs28365063) were

10

ACCEPTED MANUSCRIPT

genotyped using predesigned TaqMan SNP genotyping assays on a StepOne Plus real-time instrument (Applied Biosystems, Foster City, California, USA) in accordance with the manufacturer’s protocol. The OCT1 SNPs rs72552763 and rs34130495 were genotyped by Sanger sequencing as previously described [34]. Genotyping for the ABCB1 SNP rs1045642 was carried

PT

out using sequence specific primers and polymerase chain reaction as described in [35]. Assay numbers and sequences of the primers used for genotyping are summarized in Supplementary Table

RI

S1. The three OCT1 SNPs are all located in the coding regions of the gene and lead to amino acid

SC

changes (rs12208357 Arg-Cys, rs34130495 Gly-Ser, rs72552763 Met-Del). Furthermore, the UGT2B7 rs28365063 (Arg-Arg) and ABCB1 rs1045642 (Ala-Ser) SNPs are placed in the coding

MA

NU

regions of the gene, whereas the UGT2B7 rs7438135 SNP are not.

2.7 Statistical analysis and considerations

D

All genotype frequencies were tested for Hardy-Weinberg equilibrium using Chi-square goodness-

PT E

of-fit test (Haploview version 4.2, Broad Institute, Cambridge, USA). The demographic data are presented as medians with 25th to 75th percentile. A multivariate

CE

regression analysis was used to determine the effect of the genotypes on the pharmacokinetic parameters AUC0-150 min, ktr, CL, VD and Cmax of morphine. Additionally, the genetic associations

AC

with OCT1, UGT2B7 and ABCB1 genotypes and morphine response (Δmor – Δpla) for the pharmacodynamic variables (volume (mL), pressure (kPa) and ice AUC) were explored using multivariate regression analysis. All statistical analyses were accomplished in STATA 11.0 (StataCorp, Texas, US) and a P-value smaller than 0.05 was considered significant. P-values for the dominant (wt/wt vs. wt/v & v/v), recessive (v/v vs. wt/wt & wt/v), and additive (wt/wt vs. wt/v vs. v/v) genotype were calculated.

11

ACCEPTED MANUSCRIPT

A sample size was calculated for the pharmacodynamic outcomes of morphine for the primary endpoints [30]. However, a specific sample size calculation for the present study (secondary endpoint) was not accomplished since we aimed to include all 40 participants enrolled in the

AC

CE

PT E

D

MA

NU

SC

RI

PT

original study.

12

ACCEPTED MANUSCRIPT

3. RESULTS In the original study 39 completed the study (one male dropped out due to side effects). Two more participants were excluded due to methodical problems (no DNA from one male participant and no pharmacokinetic data from one female participant). Out of the 37 participants, 17 were women and

PT

20 men with a median age of 25 years old and these were genotyped for the ABCB1 SNP rs1045642. In the add-on study, we were in contact with 27 participants out of the 39 from the

RI

original study, but 5 rejected to participate. Hence, 22 participants from the original study gave

SC

written informed consent to provide an extra blood sample (13 women and 9 men) for the genetic association study of OCT1 and UGT2B7. Demographic characterization is shown in Table 1, and no

NU

significant differences were observed between the two groups.

MA

--- Table 1 near here --The pharmacokinetic parameters according to genotype are provided in Table 2.No significant

D

differences in the parameters were seen between the different OCT1, ABCB1, and UGT2B7

PT E

genotypes. Neither the pharmacokintics parameters obtain for M3G and M6G could not be

CE

demonstrated to vary according to genotype (all P-values > 0.05, data not presented here). --- Table 2 near here ---

AC

Morphine pharmacodynamics in the healthy volunteers according to genotype are provided in Table 3. No impact of OCT1, ABCB1, and UGT2B7 genotypes on the pharmacodynamic response to morphine could be demonstrated. --- Table 3 near here ---

13

ACCEPTED MANUSCRIPT

4. DISCUSSION Our results did not support the hypothesis that genetic variation in OCT1, ABCB1, and UGT2B7 influence morphine pharmacokinetics or -dynamics. 4.1 Methodological considerations

PT

The main weakness of this study is the explorative nature of the analysis, meaning that no sample

RI

size calculation was performed. However, previous experimental studies with a relative low number

SC

of participants have been sufficiently powered to explore the variation in pharmacokinetics between groups in comparable settings [11, 36]. Evaluating morphine plasma concentrations for only 150

NU

minutes after administration may exclude some important information about morphine metabolism and elimination. In addition, only a limited number of polymorphisms in three genes were

MA

genotyped. Thus, it cannot be excluded that other genes may affect morphine pharmacokinetics. Lastly, in genetic association studies the interaction between genes (epistasis) may complicate the

CE

4.2 OCT1

PT E

be considered for future studies.

D

analysis and interpretation. Interaction between genes were not included in this analysis but should

AC

In humans, the drug transporter OCT1 is primary localized at the basolateral membrane of epithelial cells in the liver [37] and therefore, less morphine is expected to be transported into the hepatocytes in participants with OCT1 reduced-function allele. Supporting the theory, a study has shown that in healthy Caucasian volunteers carrying one or two OCT1 reduced-function alleles a significant higher morphine AUC0-24h and a insignificant higher Cmax [11]. However, our results could neither confirm nor exclude that OCT1 reduced-function alleles have an influence on any morphine pharmacokinetic parameters.

14

ACCEPTED MANUSCRIPT

Both AUC and Cmax are highly dependent on the timing of bloodsampling, especially in studies with sparse sampling. In the study by Tzvetkov et al. [11], where higher values for AUC and Cmax were seen in volunteers with one or two OCT1 reduced-function alleles, blood samples were only collected every 30 min for the first 2 hours. The recorded Cmax was therefore the morphine

PT

concentration after 30 min for all individuals, while the actual Cmax most likely occurred before or after 30 min for most subjects. In our study, six blood samples were collected during the first 60

RI

min after administration and the absorption was described with a transit compartment constant (ktr),

SC

which was 0.226 min-1 (mean transit time = 22 min). The inter-individual variability related to the absorption parameter (ktr) was 52% CV, and thus the absorption duration varied greatly between

NU

individuals. We were not able to identify a difference in absorption rate related to OTC1

MA

polymorphism, probably due to small sample size. However, the absorption phase was much better characterised in our study than in the study by Tzvetkov et al. [11], where Cmax due to sparse

D

sampling was estimated to be the same for all subjects and thus not reflecting the true inter-

PT E

individual variability. The difference in Cmax values might therefore be related to inter-individual variability not explained by OTC1 polymorphism. The same applies for the AUC values, where the number and timing of samples is also important for an accurate estimation of the AUC. The total

CE

drug exposure might be the same for two individuals with very different absorption profiles, but if

AC

that difference in absorption is not captured due to sparse sampling, the AUC’s might be significantly different. When investigating covariate effects (including genetics) on absorption, it is crucial to have as many bloodsamples during the absorption phase as possible. Furthermore, nonlinear mixed effect modelling is superior to non-compartmental analysis as all data are analysed simultaneously to determine the typical population parameters as well as quantifying interindividual, inter-occasion, and random variability. Moreover, non-linear mixed effects modelling does not require a rich data set, which is crucial for non-compartmental analysis.

15

ACCEPTED MANUSCRIPT

4.3 ABCB1 The efflux transporter P-glycoprotein exerts its function in various tissues. It has been suggested that homozygotes of the ABCB1 3435T allele are good morphine responders whereas hetero- and homozygotes of the C allele are moderate responders [38]. Hence, it has been speculated whether

PT

the absorption of morphine is reduced in homozygotes of the C allele due to increased efflux of morphine in the gut. Consequently, reducing the bioavailability of morphine. The efflux of

RI

morphine from the gut epithelial cells is mainly expected to affect the absorption of morphine, but

SC

the transit compartment rate constant showed no significant difference between genotypes. We showed that the AUC0-150min of morphine was found slightly increased in homozygotes and

NU

heterozygotes for the 3435C allele, but these results were not statistically significant. The same

MA

trend was observed in another study of postoperative patients, where morphine concentrations 24 hours after administration were compared [39]. Thus, ABCB1 3435C allele may have a weak impact

PT E

D

on morphine pharmacokinetics, but further studies are needed to confirm this association.

4.4 UGT2B7

Glucuronidation activity may also influence the effect of morphine. The difference in AUC0-150 min

CE

was not significantly different among the UGT2B7 -900G>A (rs7438135) genotypes. Nonetheless,

AC

the trend was consistent with results from another study in patients with sickle cell disease [27], where homozygous and heterozygous carriers of the G allele had higher morphine AUC than homozygous carriers of the A allele. This indicates that the G allele may be associated with reduced glucuronidation of morphine. A rather limited number of studies have investigated morphine pharmacokinetics and the genetic influence of the UGT2B7 372A>G (rs28365063) polymorphism. Contradictory results have been published with regard to the effect of UGT2B7 polymorphism on metabolism of antiepileptic drugs [40, 41], but to our knowledge no studies exist for morphine

16

ACCEPTED MANUSCRIPT

population pharmacokinetics. The results from the present study did not provide any further evidence regarding UGT2B7 polymorphism and association to morphine pharmacokinetics.

4.5 Pain tolerance thresholds for experimental induced pain

PT

Genetic variation in genes encoding drug transporters, such as OCT1 and ABCB1, and the drug-

RI

metabolizing enzyme UGT2B7 may have an indirect influence on morphine pharmacodynamics by changing the pharmacokinetics of the drug. In the present study we were unable to demonstrate an

SC

association between these polymorphisms and the antinociceptive effect of morphine. Literature

NU

investigating this association is limited. Twin studies have shown that up to 60% of the variation in the response to experimental pain is genetically inherited [42–44]. It has been suggested that genetic

MA

variation influences different types of pain and therefore, a genetic association in one experimental

D

pain modality may not be present in another [45].

PT E

This study was explorative and we recommend for future larger scale studies to: 1) include several candidate genes e.g. OCT1, ABCB1 and UGT2B7; 2) investigate several SNPs in same candidate

CE

gene; 3) include other pain modalities (we only included pressure in multiple tissues) [46] ; 4) include test in different tissues; 5) more frequent sampling during the first hour and sampling for six

AC

hours or more to enable more precise estimation of morphine absorption and elimination; 6) analysis of the influence of genetic polymorphism using nonlinear mixed effect modelling approach on parameters such as ktr/ka, CL, and VD instead of AUC and Cmax, which allows for a more integrated analysis of the pharmacokinetic and/or pharmacodynamic data and the pharmacogenetic data than ANOVA analysis [47].

17

ACCEPTED MANUSCRIPT

4.6 Conclusion In conclusion, no statistically significant influence of OCT1, ABCB1, and UGT2B7 genotypes on morphine pharmacokinetics or pharmacodynamics could be demonstrated. Nonetheless, due to methodological limitations we cannot exclude that associations between genetic variations in OCT1,

RI

PT

ABCB1, and UGT2B7, and morphine pharmacokinetics and -dynamics exist.

SC

5. ACKNOWLEDGEMENT

The authors wish to thank laboratory technician Susanne Hillbrandt for invaluable technical

MA

NU

assistance.

6. FINANCIAL DISCLOSURE

D

This work was supported by funding from the Innovation Fund Denmark - Individuals, Disease and

AC

CE

PT E

Society. TBS has held unrelated paid lectures for Eisai, Orifarm, Novartis and Astellas-Pharma.

18

ACCEPTED MANUSCRIPT

7. REFERENCE 1.

De Gregori S, De Gregori M, Ranzani GN, Allegri M, Minella C, Regazzi M. Morphine metabolism, transport and Brain disposition. Metab Brain Dis 2012;27:1–5

2.

Cherny N, Ripamonti C, Pereira J, Davis C, Fallon M, McQuay H, Mercadante S, Pasternak

PT

G, Ventafridda V. Strategies to manage the adverse effects of oral morphine: an evidence-

Somogyi AA, Coller JK, Barratt DT. Pharmacogenetics of opioid response. Clin Pharmacol

SC

3.

RI

based report. J Clin Oncol 2001;19:2542–54

4.

NU

Ther 2015;97:125–7

Nies AT, Koepsell H, Winter S, Burk O, Klein K, Kerb R, Zanger UM, Keppler D, Schwab

MA

M, Schaeffeler E. Expression of organic cation transporters OCT1 (SLC22A1) and OCT3 (SLC22A3) is affected by genetic factors and cholestasis in human liver. Hepatology

Kerb R, Brinkmann U, Chatskaia N, Gorbunov D, Gorboulev V, Mornhinweg E, Keil A,

PT E

5.

D

2009;50:1227–40

Eichelbaum M, Koepsell H. Identification of genetic variations of the human organic cation

6.

CE

transporter hOCT1 and their functional consequences. Pharmacogenetics 2002;12:591–5 Shu Y, Leabman MK, Feng B, Mangravite LM, Huang CC, Stryke D, Kawamoto M, Johns

AC

SJ, DeYoung J, Carlson E, Ferrin TE, Herskowitz I, Giacomini KM. Evolutionary conservation predicts function of variants of the human organic cation transporter, OCT1. Proc Natl Acad Sci U S A 2003;100:5902–7 7.

Shu Y, Sheardown SA, Brown C, Owen RP, Zhang S, Castro RA, Lanculescu AG, Yue L, Lo JC, Burchard EG, Brett CM, Giacomini KM. Effect of genetic variation in the organic cation transporter 1 (OCT1) on metformin action. J Clin Invest 2007;117:1422–31

19

ACCEPTED MANUSCRIPT

8.

Tzvetkov M V, Saadatmand AR, Bokelmann K, Meineke I, Kaiser R, Brockmöller J. Effects of OCT1 polymorphisms on the cellular uptake, plasma concentrations and efficacy of the 5HT3 antagonists tropisetron and ondansetron. Pharmacogenomics J 2010;12:22–29

9.

Tzvetkov M V, Saadatmand AR, Lötsch J, Tegeder I, Stingl JC, Brockmöller J. Genetically

PT

Polymorphic OCT1: Another Piece in the Puzzle of the Variable Pharmacokinetics and Pharmacodynamics of the Opioidergic Drug Tramadol. Clin Pharmacol Ther 2011;90:143–

Ahlin G, Karlsson J, Pedersen

, ustavsson L, Larsson ,

SC

10.

RI

150

atsson , orinder U,

NU

ergstr m CA , Artursson P. Structural Requirements for Drug Inhibition of the Liver Specific Human Organic Cation Transport Protein 1. J Med Chem 2008;51:5932–5942 Tzvetkov M V, dos Santos Pereira JN, Meineke I, Saadatmand AR, Stingl JC, Brockmöller J.

MA

11.

Morphine is a substrate of the organic cation transporter OCT1 and polymorphisms in OCT1

12.

PT E

2013;86:666–78

D

gene affect morphine pharmacokinetics after codeine administration. Biochem Pharmacol

Thiebaut F, Tsuruo T, Hamada H, Gottesman MM, Pastan I, Willingham MC. Cellular

CE

localization of the multidrug-resistance gene product P-glycoprotein in normal human

13.

AC

tissues. Proc Natl Acad Sci U S A 1987;84:7735–8 Thompson SJ, Koszdin K, Bernards CM. Opiate-induced analgesia is increased and prolonged in mice lacking P-glycoprotein. Anesthesiology 2000;92:1392–9 14.

Ishikawa T, Hirano H, Onishi Y, Sakurai A, Tarui S. Functional evaluation of ABCB1 (Pglycoprotein) polymorphisms: high-speed screening and structure-activity relationship analyses. Drug Metab Pharmacokinet 2004;19:1–14

15.

Kimchi-Sarfaty C, Oh JM, Kim I-W, Sauna ZE, Calcagno AM, Ambudkar S V, Gottesman 20

ACCEPTED MANUSCRIPT

MM. A “silent” polymorphism in the

D 1 gene changes substrate specificity. Science

2007;315:525–8 16.

Sakaeda T, Nakamura T, Okumura K. Pharmacogenetics of MDR1 and its impact on the pharmacokinetics and pharmacodynamics of drugs. Pharmacogenomics 2003;4:397–410 Hoffmeyer S, Burk O, von Richter O, Arnold HP, Brockmöller J, Johne A, Cascorbi I,

PT

17.

RI

Gerloff T, Roots I, Eichelbaum M, Brinkmann U. Functional polymorphisms of the human

SC

multidrug-resistance gene: multiple sequence variations and correlation of one allele with Pglycoprotein expression and activity in vivo. Proc Natl Acad Sci U S A 2000;97:3473–8 Hitzl M, Drescher S, van der Kuip H, Schäffeler E, Fischer J, Schwab M, Eichelbaum M,

NU

18.

Fromm MF. The C3435T mutation in the human MDR1 gene is associated with altered

Siegsmund M, Brinkmann U, Scháffeler E, Weirich G, Schwab M, Eichelbaum M, Fritz P,

PT E

19.

D

Pharmacogenetics 2001;11:293–8

MA

efflux of the P-glycoprotein substrate rhodamine 123 from CD56+ natural killer cells.

Burk O, Decker J, Alken P, Rothenpieler U, Kerb R, Hoffmeyer S, Brauch H. Association of the P-glycoprotein transporter MDR1(C3435T) polymorphism with the susceptibility to renal

Xie R, Hammarlund-Udenaes M, de Boer AG, de Lange EC. The role of P-glycoprotein in

AC

20.

CE

epithelial tumors. J Am Soc Nephrol 2002;13:1847–54

blood-brain barrier transport of morphine: transcortical microdialysis studies in mdr1a (-/-) and mdr1a (+/+) mice. Br J Pharmacol 1999;128:563–8 21.

King M, Su W, Chang A, Zuckerman A, Pasternak GW. Transport of opioids from the brain to the periphery by P-glycoprotein: peripheral actions of central drugs. Nat Neurosci 2001;4:268–74

22.

Lötsch J, Schmidt R, Vetter G, Schmidt H, Niederberger E, Geisslinger G, Tegeder I. 21

ACCEPTED MANUSCRIPT

Increased CNS uptake and enhanced antinociception of morphine-6-glucuronide in rats after inhibition of P-glycoprotein. J Neurochem 2002;83:241–8 23.

Coffman BL, Rios GR, King CD, Tephly TR. Human UGT2B7 catalyzes morphine glucuronidation. Drug Metab Dispos 1997;25:1–4 Bhasker CR, McKinnon W, Stone A, Lo AC, Kubota T, Ishizaki T, Miners JO. Genetic

PT

24.

RI

polymorphism of UDP-glucuronosyltransferase 2B7 (UGT2B7) at amino acid 268: ethnic

25.

SC

diversity of alleles and potential clinical significance. Pharmacogenetics 2000;10:679–85 Innocenti F, Liu W, Fackenthal D, Ramírez J, Chen P, Ye X, Wu X, Zhang W, Mirkov S,

NU

Das S, Cook Jr. E, Retain M. Single nucleotide polymorphism discovery and functional assessment of variation in the UDP-glucuronosyltransferase 2B7 gene. Pharmacogenet

Matic M, Norman E, Rane A, Beck O, Andersson M, Elens L, Tibboel D, Fellman V, van

D

26.

MA

Genomics 2008;18:683–97

PT E

Schaik RHN. Effect of UGT2B7 -900G>A (-842G>A; rs7438135) on morphine glucuronidation in preterm newborns: results from a pilot cohort. Pharmacogenomics

27.

CE

2014;15:1589–97

Darbari DS, van Schaik RHN, Capparelli E V, Rana S, McCarter R, van den Anker J.

AC

UGT2B7 promoter variant -840G>A contributes to the variability in hepatic clearance of morphine in patients with sickle cell disease. Am J Hematol 2008;83:200–2 28.

Edwards RR, Sarlani E, Wesselmann U, Fillingim RB. Quantitative assessment of experimental pain perception: multiple domains of clinical relevance. Pain 2005;114:315–9

29.

Arendt-Nielsen L, Yarnitsky D. Experimental and Clinical Applications of Quantitative Sensory Testing Applied to Skin, Muscles and Viscera. J Pain 2009;10:556–572

22

ACCEPTED MANUSCRIPT

30.

Olesen AE, Brock C, Sverrisdóttir E, Larsen IM, Drewes AM. Sensitivity of quantitative sensory models to morphine analgesia in humans. J Pain Res 2014;7:717–26

31.

Sverrisdóttir E, Foster DJR, Upton RN, Olesen AE, Lund TM, Gabel-Jensen C, Drewes AM, Christrup LL, Kreilgaard M. Modelling concentration-analgesia relationships for morphine to

PT

evaluate experimental pain models. Eur J Pharm Sci 2014;66C:50–58 Team RDC tR: A language and environment for statistical computing. 2005;3

33.

Drewes AM, Gregersen H, Arendt-Nielsen L. Experimental pain in gastroenterology: a

SC

RI

32.

reappraisal of human studies. Scand J Gastroenterol 2003;38:1115–30 Christensen MMH, Brasch-Andersen C, Green H, Nielsen F, Damkier P, Beck-Nielsen H,

NU

34.

MA

Brosen K. The pharmacogenetics of metformin and its impact on plasma metformin steadystate levels and glycosylated hemoglobin A1c. Pharmacogenet Genomics 2011;21:837–50 Ross JR, Riley J, Taegetmeyer AB, Sato H, Gretton S, du Bois RM, Welsh KI. Genetic

D

35.

PT E

variation and response to morphine in cancer patients: catechol-O-methyltransferase and multidrug resistance-1 gene polymorphisms are associated with central side effects. Cancer

36.

CE

2008;112:1390–403

Christensen MMH, Højlund K, Hother-Nielsen O, Stage TB, Damkier P, Beck-Nielsen H,

AC

Brøsen K. Steady-state pharmacokinetics of metformin is independent of the OCT1 genotype in healthy volunteers. Eur J Clin Pharmacol 2015;71:691–7 37.

Gorboulev V, Ulzheimer JC, Akhoundova A, Ulzheimer-Teuber I, Karbach U, Quester S, Baumann C, Lang F, Busch AE, Koepsell H. Cloning and characterization of two human polyspecific organic cation transporters. DNA Cell Biol 1997;16:871–81

38.

Campa D, Gioia A, Tomei A, Poli P, Barale R. Association of ABCB1/MDR1 and OPRM1

23

ACCEPTED MANUSCRIPT

gene polymorphisms with morphine pain relief. Clin Pharmacol Ther 2008;83:559–66 39.

Bastami S, Gupta A, Zackrisson A-L, Ahlner J, Osman A, Uppugunduri S. Influence of UGT2B7, OPRM1 and ABCB1 gene polymorphisms on postoperative morphine consumption. Basic Clin Pharmacol Toxicol 2014;115:423–31 Puranik YG, Birnbaum AK, Marino SE, Ahmed G, Cloyd JC, Remmel RP, Leppik IE,

PT

40.

RI

Lamba JK. Association of carbamazepine major metabolism and transport pathway gene

SC

polymorphisms and pharmacokinetics in patients with epilepsy. Pharmacogenomics 2013;14:35–45

Singkham N, Towanabut S, Lertkachatarn S, Punyawudho B. Influence of the UGT2B7 -

NU

41.

161C>T polymorphism on the population pharmacokinetics of lamotrigine in Thai patients.

Angst MS, Phillips NG, Drover DR, Tingle M, Ray A, Swan GE, Lazzeroni LC, Clark JD.

D

42.

MA

Eur J Clin Pharmacol 2013;69:1285–1291

PT E

Pain sensitivity and opioid analgesia: a pharmacogenomic twin study. Pain 2012;153:1397– 409

Nielsen CS, Stubhaug A, Price DD, Vassend O, Czajkowski N, Harris JR. Individual

CE

43.

differences in pain sensitivity: genetic and environmental contributions. Pain 2008;136:21–9 Norbury T a., MacGregor AJ, Urwin J, Spector TD, McMahon SB. Heritability of responses

AC

44.

to painful stimuli in women: A classical twin study. Brain 2007;130:3041–3049 45.

Sato H, Droney J, Ross J, Olesen AE, Staahl C, Andresen T, Branford R, Riley J, ArendtNielsen L, Drewes AM. Gender, variation in opioid receptor genes and sensitivity to experimental pain. Mol Pain 2013;9:20

24

ACCEPTED MANUSCRIPT

46.

Olesen AE, Sato H, Nielsen LM, Staahl C, Droney J, Gretton S, Branford R, Drewes AM, Arendt-Nielsen L, Riley J, Ross J. The genetic influences on oxycodone response characteristics in human experimental pain. Fundam Clin Pharmacol 2015;29:417–425 Joerger M. Covariate Pharmacokinetic Model Building in Oncology and its Potential Clinical

CE

PT E

D

MA

NU

SC

RI

PT

Relevance. The AAPS Journal 2012;14:119-32

AC

47.

25

ACCEPTED MANUSCRIPT

TABLES Table 1 Demographic characterization of the original study (n=37) and the present study (n=22) Demographic characteritics

Number

Median

25-75 percentile

Age at inclusion (years)

25

24-26

Height (m)

1.8

1.74-1.85

Weight (kg)

78

72-87

BMI (kg/m2)

24.7

22.8-26.3

Age at inclusion (years)

25

25-26.8

Height (m)

1.8

1.73-1.85

78.5

72.3-87

Original study (n=37):

Present study (n=22):

Weight (kg) 2

BMI (kg/m )

25.1

23.7-26.1

2

AC

CE

PT E

D

MA

BMI: Body Mass Index=weight/height

SC

13:9

NU

Gender (women:men)

PT

17:20

RI

Gender (women:men)

26

ACCEPTED MANUSCRIPT Table 2 Median with 25th and 75th percentile trough pharmacokinetics of the healthy volunteers treated with morphine according to genotype Gene OCT1 (n=22)

ABCB1 (n=37)

dbSNP Diplotypes for RF

rs1045642 (wt=T)

n

wt/wt Median(25-75th percentile)

AUC0-150min (min* ng/L)

14

1395 (1177 to 1502)

5

1180 (915 to 1582)

3

ktr (min-1)

14

0.2 (0.1 to 0.3)

5

0.2 (0.1 to 0.3)

3

CL (L/min)

14

17 (14.5 to 20.9)

5

17.7 (17 to 27.5)

3

VD (L)

14

1421 (1000 to 1683)

5

1933 (918 to 2054)

Cmax (ng/ml)

14

15.7 (13.2 to 20.2)

5

AUC0-150min (min* ng/L)

12

1500 (1121 to 1702)

20

ktr (min-1)

12

0.2 (0.2 to 0.3)

M

CL (L/min)

12

VD (L)

12

Parameter

UGT2B7 (n=22)

rs7438135 (wt=A)

12

n

v/v Median(25-75th percentile)

Pvalues

1312 (1216 to 1597)

0.69

T P

I R

0.2 (0.1 to 0.4)

0.72

17 (11.2 to 18.1)

0.40

3

1650 (875 to 1659)

0.61

13.1 (11.3 to 22.4)

3

15 (13.7 to 17.5)

0.65

1398 (1200 to 1624)

5

1311 (1229 to 1582)

0.51

20

0.2 (0.1 to 0.3)

5

0.3 (0.2 to 0.3)

0.91

16 (11 to 20.8)

20

16.2 (13.1 to 19.3)

5

17 (17 to 17.7)

0.83

1223 (831 to 1835)

20

1406 (970 to 1706)

5

1398 (1265 to 1658)

0.95

17.7 (12.8 to 22)

20

15.7 (13.4 to 19.5)

5

15.2 (15.0 to 18.1)

0.43

D E

PT

E C

Cmax (ng/ml)

n

wt/v Median(25-75th percentile)

C S U

N A

AUC0-150min (min* ng/L)

2

1129 (1042 to 1216)

15

1448 (940 to 1597)

5

1341 (1275 to 1489)

0.32

ktr (min-1)

2

0.3 (0.2 to 0.3)

15

0.2 (0.1 to 0.3)

5

0.2 (0.1 to 0.3)

0.93

CL (L/min)

2

21.3 (18.1 to 24.5)

15

17.2 (15.2 to 23.6)

5

14.5 (12.8 to 15.5)

0.47

VD (L)

2

1633 (1616 to 1650)

15

1113 (918 to 1933)

5

1490 (1352 to 1683)

0.63

Cmax (ng/ml)

2

14 (13.7 to 14.3)

15

17.5 (12.3 to 22.4)

5

14.7 (13.2 to 16.7)

0.45

C A

27

ACCEPTED MANUSCRIPT rs28365063 (wt=A)

AUC0-150min (min* ng/L)

15

1448 (1184 to 1582)

7

1180 (940 to 1498)

0

-

0.15

ktr (min-1)

15

0.2 (0.1 to 0.4)

7

0.3 (0.2 to 0.3)

0

-

0.92

CL (L/min)

15

15.6 (13.6 to 18.8)

7

18.1 (17.2 to 27)

0

-

0.05

VD (L)

15

1352 (930 to 1683)

7

1650 (1095 to 1933)

0

Cmax (ng/ml)

15

16.7 (13.2 to 22.4)

7

13.7 (12.3 to 18.6)

0

I R

T P -

0.40

-

0.40

Diplotypes for RF: reduced-function of the SNPs rs12208357, rs34130495 and rs72552763, dbSNP: single nucleotide polymorphism database identification, wt = wildtype, v = variant, P-values are for the dominant genotype (wt/wt vs. wt/v & v/v) (P-values for the recessive and additive genotype are not shown – all P > 0.05),

C S U

AUC0-150min = area under the plasma concentration-time curve from morphine administration to 150 minutes after, ktr= transit compartment rate constant, CL = clearance, VD = volume of distribution, Cmax = the maximum plasma concentration

N A

D E

M

T P E

C C

A

28

ACCEPTED MANUSCRIPT Table 3 Median with 25th and 75th percentile trough pharmacodynamics of the healthy volunteers treated with morphine according to genotype wt/wt Gene

dbSNP

OCT1 (n=22)

Diplotypes for RF

ABCB1 (n=37)

rs1045642 (wt=T)

Parameter

rs7438135 (wt=A)

v/v Median(25-75th percentile)

P-values

2

8.7 (2.5 to 14.8)

0.75

3

1.3 (-2.9 to 6.7)

0.82

-8.9 (-17.3 to -3)

3

-4.5 (-11.6 to -0.5)

0.32

19

4.4 (-15.3 to 32.9)

4

38.5 (13.8 to 72.3)

0.85

13.2 (-3.8 to 33.2)

20

13.4 (22.0 to 28.1)

5

1.3 (-26.4 to 6.3)

0.94

12

-9.1 (-14.5 to -1.7)

17

-7.0 (-16.3 to -2.0)

3

-3.5 (-8.8 to 0.4)

0.81

2

2.4 (-10 to 14.8)

14

25.9 (2.5 to 47.3)

5

-8.5 (-18.5 to -4.4)

0.92

Muscle pressure stimulation (PP)

2

8.7 (6.7 to 10.7)

15

1.3 (-12.7 to 40.8)

5

34.9 (21.7 to 43.1)

0.71

ice AUC for cold pressor test (PP)

2

-5.3 (-17.6 to 6.9)

11

-3.6 (-7.1 to -0.5)

5

-8.0 (-11.6 to -0.9)

0.98

n Rectal pressure stimulation (PP)

14

3.4 (-10 to 44.5)

5

25.6 (3.5 to 51.7)

Muscle pressure stimulation (PP)

14

23.1 (-4.7 to 40.8)

5

-5.3 (-24.6 to 43.9)

ice AUC for cold pressor test (PP)

13

-5.3 (-8 to 1.3)

2

Rectal pressure stimulation (PP)

12

1.43 (-7.2 to 16.1)

Muscle pressure stimulation (PP)

12

C C

A

Median(25-75 percentile)

D E

M

n

T P

I R

C S U

N A

T P E

Rectal pressure stimulation (PP)

n

th

Median(25-75 percentile)

ice AUC for cold pressor test (PP) UGT2B7 (n=22)

wt/v th

29

ACCEPTED MANUSCRIPT rs28365063 (wt=A)

Rectal pressure stimulation (PP)

14

15.8 (-8.5 to 44.5)

7

3.5 (-10.0 to 66.3)

0

-

0.82

Muscle pressure stimulation (PP)

15

11.1 (-12.7 to 43.1)

7

10.7 (-24.6 to 29.4)

0

-

0.53

ice AUC for cold pressor test (PP)

14

-6.2 (-11.6 to -0.9)

4

-6.2 (-16.6 to 4.4)

-

0.99

I R

T P 0

All dynamic variables are presented as morphine response (Δmorphine – Δplacebo) presented as percentage points ( ) where Δmorphine and Δplacebo are calculated as percentage change from baseline ((T60 min-baseline)/baseline*100%), Diplotypes for RF: reduced-function of the SNPs rs12208357, rs34130495 and rs72552763, dbSNP: single nucleotide polymorphism database identification, wt = wildtype, v = variant, P-values are for the dominant genotype (wt/wt vs. wt/v & v/v) (P-values for the recessive and additive genotype are not shown – all P > 0.05), ice AUC for the cold pressor test: area under time-pain level curve for the cold pressor test.

C S U

N A

D E

M

T P E

C C

A

30

AC

CE

PT E

D

MA

NU

SC

RI

PT

ACCEPTED MANUSCRIPT

Graphical abstract

31